Neurovascular unit

Last updated

The neurovascular unit (NVU) comprises the components of the brain that collectively regulate cerebral blood flow in order to deliver the requisite nutrients to activated neurons. [1] The NVU addresses the brain's unique dilemma of having high energy demands yet low energy storage capacity. In order to function properly, the brain must receive substrates for energy metabolism–mainly glucose–in specific areas, quantities, and times. [2] Neurons do not have the same ability as, for example, muscle cells, which can use up their energy reserves and refill them later; therefore, cerebral metabolism must be driven in the moment. The neurovascular unit facilitates this ad hoc delivery and, thus, ensures that neuronal activity can continue seamlessly. [2]

Contents

The neurovascular unit was formalized as a concept in 2001, at the inaugural Stroke Progress Review Group of the National Institute of Neurological Disorders and Stroke (NINDS). [1] In prior years, the importance of both neurons and cerebral vasculature was well known; however, their interconnected relationship was not. The two were long considered distinct entities which, for the most part, operated independently. Since 2001, though, the rapid increase of scientific papers citing the neurovascular unit represents the growing understanding of the interactions that occur between the brain’s cells and blood vessels. [1]

The neurovascular unit consists of neurons, astrocytes, vasculature (endothelial and vascular mural cells), the vasomotor apparatus (smooth muscle cells and pericytes), and microglia. [1] Together these function in the homeostatic haemodynamic response of cerebral hyperaemia. [3] Cerebral hyperaemia is a fundamental central nervous system mechanism of homeostasis that increases blood supply to neural tissue when necessary. [3] This mechanism controls oxygen and nutrient levels using vasodilation and vasoconstriction in a multidimensional process involving the many cells of the neurovascular unit, along with multiple signaling molecules. [1] The interactions between the components of the NVU allow it to sense neurons' needs of oxygen and glucose and, in turn, trigger the appropriate vasodilatory or vasoconstrictive responses. [3] Neuronal activity [4] as well as astrocytes [5] can therefore participate in CNV, both by inducing vasodilation and vasoconstriction [6] .Thus, the NVU provides the architecture behind neurovascular coupling, which connects neuronal activity to cerebral blood flow and highlights the interdependence of their development, structure, and function. [1]

A schematic of the neurovascular unit (NVU), where astrocyte processes surround the capillary basement membrane and pericytes, creating the glia limitans. Also, resident in the perivascular space are antigen-presenting cells (APCs) and border-associated macrophages (BAMs). Neurovascular unit.jpg
A schematic of the neurovascular unit (NVU), where astrocyte processes surround the capillary basement membrane and pericytes, creating the glia limitans. Also, resident in the perivascular space are antigen-presenting cells (APCs) and border-associated macrophages (BAMs).

The temporal and spatial link between cerebral blood flow and neuronal activity allows the former to serve as a proxy for the latter. Neuroimaging techniques that directly or indirectly monitor blood flow, such as fMRI and PET scans, can, thus, measure and locate activity in the brain with precision. [1] Imaging of the brain also allows researchers to better understand the neurovascular unit and its many complexities. Furthermore, any impediments to the function of the neurovascular system will prevent neurons from receiving the appropriate nutrients. A complete stoppage for only a few minutes, which could be caused by arterial occlusion or heart failure, can result in permanent damage and death. Dysfunction in the NVU is also associated with neurodegenerative diseases including Alzheimer's and Huntington's disease. [1]

Function

Anatomical components

The neurovascular unit is made up of vascular cells (including endothelium, pericytes, and smooth muscle cells), glia (astrocytes and microglia), and neurons with synaptic junctions for signaling. [1] Cerebral vessels, namely arterioles and the perivascular compartment, form the network of the NVU. [7] Arterioles are made up of pial vessels and arterioles, and the perivascular compartment includes perivascular macrophages in addition to Mato, pial, and mast cells. Cerebral blood flow is a critical component of this overall system and it is facilitated by the neck arteries. Segmented vascular resistance, or the amount of flow control that each section of the brain maintains, is measured as the ratio of the blood pressure gradient to blood flow volume. [8] The blood flow within the NVU is a low resistance channel that allows blood to be distributed to different parts of the body. [9] The cells of the NVU sense the needs of neural tissue and release many different mediators that engage in signaling pathways and initiate effector systems such as the myogenic effect; these mediators trigger the vascular smooth muscle cells to increase blood flow through vasodilation or to reduce blood flow by vasoconstriction. [3] [1] [10] This is recognized as a multidimensional response that operates across the cerebrovascular network as a whole. [1]

Blood–brain barrier

The cells of the neurovascular unit also make up the blood–brain barrier (BBB), which plays an important role in maintaining the microenvironment of the brain. [11] In addition to regulating the exit and entrance of blood, the blood–brain barrier also filters toxins that may cause inflammation, injury, and disease. [12] The overall microvasculature unit functions as a defense for the central nervous system. [11] Encompassed within the BBB are two types of blood vessels: endothelial and mural cells. Endothelial cells form the wall of the BBB, while mural cells exist on the outer surface of this layer of endothelial cells. The mural cells also have their own abluminal layer which hosts pericytes that work to maintain the permeability of the barrier, and the epithelial cells filter the amount of toxins entering. These cells connect to different segments of the vascular tree that exist within the brain. [12]

Neurovascular coupling

Cellular processes critically rely on the production of adenosine triphosphate (ATP), which requires glucose and oxygen. [13] These need to be delivered to areas in the brain with consistency via cerebral blood flow. In order for the brain to receive enough blood flow when in high demand, coupling occurs between neurons and CBF. Neurovascular coupling encompasses the changes in cerebral blood flow that occur in response to the level of neuronal activity. [1] [14] [15] When the brain needs to exert more energy, there is an associated increase in the level of blood flow to compensate for this. The brain does not have a place where it stores energy, and, therefore, the response of blood flow has to be immediate so that crucial functions for continued life can persist. Difficulties arise when angiotensin proteins are present in higher concentrations, as there is an associated increase in blood flow that leads to hypertension and potential disorders. [8] Furthermore, modern imaging techniques have allowed researchers to view and study cerebral blood flow in a noninvasive manner. However, imaging deep brain structures in vivo is challenging. Therefore, NVC can be studied on ex vivo brain slices maintained in survival conditions. [16] [17] Ultimately, neurovascular coupling promotes brain health by moderating proper cerebral blood flow. There is still much more to be discovered about it, though; and, due to the difficulty of in vivo research, the growing body of knowledge on neurovascular coupling relies heavily on ex vivo techniques for imaging the neurovascular unit.

Imaging

The neurovascular unit enables imaging techniques to measure neuronal activity by tracking blood flow. Various other types of neuroimaging also allow the NVU itself to be studied by providing visual insights into the complex interactions between neurons, glial cells, and blood vessels in the brain.

Fluorescence microscopy

Fluorescence microscopy is a widely used imaging technique that utilizes fluorescent probes to visualize specific molecules or structures within the neurovascular unit. [18] It allows researchers to label and track cellular components, such as neurons, astrocytes, and blood vessel markers, with high specificity. [19] Fluorescence imaging offers excellent spatial resolution, allowing for detailed visualization of cellular morphology and localized molecular interactions. [20] By using different fluorophores, researchers can simultaneously examine multiple cellular components and molecular pathways of the neurovascular unit. However, limited tissue penetration depth, photobleaching, and phototoxicity negatively impact the potential for long-term imaging studies. [20]

Electron microscopy

Electron microscopy provides details of the neurovascular unit at the nanometer scale by using a focused beam of electrons instead of light, enabling higher resolution imaging. Transmission electron microscopy images thin tissue sections, providing detailed information about the fine cellular structures, including synapses and organelles. [21] Scanning electron microscopy, on the other hand, provides 3D information by scanning a focused electron beam across the sample's surface, allowing for the visualization of the topography of neurovascular unit components. [22] Electron microscopy techniques are, thus, invaluable for studying the precise cellular and subcellular interactions within the NVU. [23] However, it requires sample preparation involving fixation, dehydration, and staining, which can introduce artifacts, and it is not suitable for live or large-scale imaging due to its time-consuming nature.

Magnetic resonance imaging

Magnetic resonance imaging (MRI) is a non-invasive imaging technique that uses strong magnetic fields and radio waves to generate detailed images of the brain's anatomy and function. [24] It can provide information about blood flow, oxygenation levels, and structural characteristics of the neurovascular unit. The functional MRI (fMRI) allows researchers to study brain activity by measuring changes in blood oxygenation associated with neural activity, thus classifying it as a blood-oxygen-level-dependent imaging (BOLD imaging) technique. Diffusion MRI (dMRI) provides insights into the brain's structural connectivity by tracking the diffusion of water molecules in its tissue. [25] MRI, in general, has excellent spatial resolution and can be used for both human and animal studies, making it a valuable tool for studying the neurovascular unit in vivo. It has limited temporal resolution, though, and its ability to visualize finer cellular and molecular details within the neurovascular unit is relatively lower compared to microscopy techniques.

Optical coherence tomography

Optical coherence tomography (OCT) is an imaging technique that utilizes low-coherence interferometry to generate high-resolution cross-sectional images of biological tissues. [26] It can, thus, provide information about the microstructure and vascular network of the neurovascular unit. [27] More specifically, OCT has been used to study cerebral blood flow dynamics, changes in vessel diameter, and blood–brain barrier integrity. It also has real-time imaging capabilities and can, thus, be effectively applied in both clinical and preclinical settings. [27] Downsides of optical coherence tomography include limited depth penetration in highly scattering tissues and a lower resolution in increasing depth, which can limit its application in deep brain regions. [26]

Clinical significance

Neurovascular failure

Neurovascular failure, or neurovascular disease, refers to a range of conditions that negatively affect the function of blood vessels in the brain and spinal cord. [28] While the exact mechanisms behind neurovascular disease are unknown, people with inherited conditions (such as a family history of heart disease, diabetes, and/or high cholesterol), poor lifestyle choices, genetic changes during pregnancy, physical trauma, and other specific genetic characteristics are generally at higher risk. [28] In particular, neurovascular failure can be caused by problems arising in the blood vessels, including blockages (embolism), clot formation (thrombosis), narrowing (stenosis), and rupture (hemorrhage). In response to pathogenic stimuli, such as tissue hypoxia, signaling pathways involved in neurovascular coupling are impaired. [29] [30] Neuronal injury is often preceded by the expression and release of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF); in addition to this, the upregulation of astrocyte receptors in endothelial cells can stimulate endothelial proliferation and migration, which can dangerously increase blood–brain barrier (BBB) permeability. [29] Ultimately, vascular dysfunction results in decreased cerebral blood flow and abnormalities in the blood–brain barrier, which poses a threat to the normal functioning of the brain. [31]

Effects of neurovascular failure

Efficient blood supply to the brain is extremely significant to its normal functioning, and improper blood flow can lead to potentially devastating neurological consequences. [31] Alterations of vascular regulatory mechanisms lead to brain dysfunction and disease. The emerging view is that neurovascular dysfunction is a feature not only of cerebrovascular pathologies, such as stroke, but also of neurodegenerative conditions, such as Alzheimer's disease. [32] While studies are still ongoing to determine the precise effects of neurovascular failure, there is emerging evidence that neurovascular dysfunction plays a pivotal role in the degeneration of the nervous system, which contrasts the typical view that neurodegeneration is caused by intrinsic neuronal effects. [29] [33] The breakdown of neurovascular coupling (e.g., modulations in neuronal activity that cause changes in local blood flow [8] ) and the pathophysiology of the NVU is commonly observed across a wide variety of neurological and psychiatric disorders, including Alzheimer’s disease. [29] The combination of recent hypotheses and evidence suggests that the pathophysiology of the NVU may contribute to cognitive impairment and be an initiating trigger for neurological manifestations of diseases such as Alzheimer's and dementia. [34] [30] Ultimately, despite the vast amount of current literature supporting vascular contributions to neurological phenotypes, there is still much to be investigated, especially with respect to the effect of neurovasculature on neurological diseases; namely, whether the initiating event occurs at the neuronal level and "mobilizes" vascular response or the vascular event triggers neuronal dysfunction. [29] [30]

Alzheimer's disease

Alzheimer's disease (AD) is the most common type of dementia, a neurodegenerative disease with progressive impairment of behavioral and cognitive functions. [35] Neuropathologically, there are two major indicators of Alzheimer's: neurofibrillary tangles (NFTs) and an accumulation of amyloid β peptide (Aβ) in the brain, known as amyloid plaques, or around blood vessels, known as amyloid angiopathy. [36] There is growing support for the vascular hypothesis of AD, which posits that blood vessels are the origin for a variety of pathogenic pathways that lead to neuronal damage and AD. [37] Vascular risk factors can result in dysregulation of the neurovascular unit and hypoxia. Destruction of the organization of the blood–brain barrier, decreased cerebral blood flow, and the establishment of an inflammatory context often result in neuronal damage since these factors promote the aggregation of β-amyloid peptide in the brain. [37] During a review of various consortium data, it was shown that more than 30% of AD cases exhibit cerebrovascular disease on post-mortem examination, and almost all have evidence of cerebral amyloid angiopathy, microvascular degeneration, and white matter lesions. [38] Despite this data, it is still insufficient to reach a pathologic diagnosis, making it unclear whether AD is a cause or a consequence of neuronal dysfunction. [32] [37] However, considering that AD seems to include a combination of vascular and neurodegenerative processes and that disruption to the vascular physiology occurs early in the disease process, targeting the vascular component may help potentially decelerate the pathologic progression of AD. [39] Currently, only a few vascular targets have been the subject of large-scale randomised controlled trials. [39]

Huntington's disease

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by an abnormal repetition of the CAG trinucleotide repeat within the Huntingtin gene (Htt). [40] Common features of Huntington's include involuntary movements (chorea), bradykinesia, psychiatric symptoms, and cognitive decline, all of which are accelerated through neuronal cell death. [41] [42] The idea that neurovascular impairments may contribute to early neuronal cell loss in Huntington’s disease has been attracting significant attention in the HD community. Reduced cerebral blood flow, increased small vessel density, and increased blood–brain barrier (BBB) permeability–all traits of neurovascular dysfunction–have been reported in both rodent and patient post-mortem tissue. [43] [44] [45] Preliminary findings support that neurovascular alterations occur in Huntington's disease and may contribute to its early neuropathology. [46] It has also been proposed that neurovascular dysregulation manifests earlier in Huntington's than other pathologies, triggering innate immune signaling and a reduction of protein levels critical for maintaining the blood–brain barrier. [47] While neurovascular failure in HD pathogenesis is still being tested, recent work supports clinical application. For example, immunohistological assays revealed vessel aberrations in brain tissue, establishing the early onset of such aberrations as a potential biomarker for early Huntington's diagnosis. [33]

Related Research Articles

<span class="mw-page-title-main">Haemodynamic response</span>

In haemodynamics, the body must respond to physical activities, external temperature, and other factors by homeostatically adjusting its blood flow to deliver nutrients such as oxygen and glucose to stressed tissues and allow them to function. Haemodynamic response (HR) allows the rapid delivery of blood to active neuronal tissues. The brain consumes large amounts of energy but does not have a reservoir of stored energy substrates. Since higher processes in the brain occur almost constantly, cerebral blood flow is essential for the maintenance of neurons, astrocytes, and other cells of the brain. This coupling between neuronal activity and blood flow is also referred to as neurovascular coupling.

<span class="mw-page-title-main">Astrocyte</span> Type of brain cell

Astrocytes, also known collectively as astroglia, are characteristic star-shaped glial cells in the brain and spinal cord. They perform many functions, including biochemical control of endothelial cells that form the blood–brain barrier, provision of nutrients to the nervous tissue, maintenance of extracellular ion balance, regulation of cerebral blood flow, and a role in the repair and scarring process of the brain and spinal cord following infection and traumatic injuries. The proportion of astrocytes in the brain is not well defined; depending on the counting technique used, studies have found that the astrocyte proportion varies by region and ranges from 20% to around 40% of all glia. Another study reports that astrocytes are the most numerous cell type in the brain. Astrocytes are the major source of cholesterol in the central nervous system. Apolipoprotein E transports cholesterol from astrocytes to neurons and other glial cells, regulating cell signaling in the brain. Astrocytes in humans are more than twenty times larger than in rodent brains, and make contact with more than ten times the number of synapses.

<span class="mw-page-title-main">Astrogliosis</span> Increase in astrocytes in response to brain injury

Astrogliosis is an abnormal increase in the number of astrocytes due to the destruction of nearby neurons from central nervous system (CNS) trauma, infection, ischemia, stroke, autoimmune responses or neurodegenerative disease. In healthy neural tissue, astrocytes play critical roles in energy provision, regulation of blood flow, homeostasis of extracellular fluid, homeostasis of ions and transmitters, regulation of synapse function and synaptic remodeling. Astrogliosis changes the molecular expression and morphology of astrocytes, in response to infection for example, in severe cases causing glial scar formation that may inhibit axon regeneration.

<span class="mw-page-title-main">Pericyte</span> Cells associated with capillary linings

Pericytes are multi-functional mural cells of the microcirculation that wrap around the endothelial cells that line the capillaries throughout the body. Pericytes are embedded in the basement membrane of blood capillaries, where they communicate with endothelial cells by means of both direct physical contact and paracrine signaling. The morphology, distribution, density and molecular fingerprints of pericytes vary between organs and vascular beds. Pericytes help to maintain homeostatic and hemostatic functions in the brain, one of the organs with higher pericyte coverage, and also sustain the blood–brain barrier. These cells are also a key component of the neurovascular unit, which includes endothelial cells, astrocytes, and neurons. Pericytes have been postulated to regulate capillary blood flow and the clearance and phagocytosis of cellular debris in vitro. Pericytes stabilize and monitor the maturation of endothelial cells by means of direct communication between the cell membrane as well as through paracrine signaling. A deficiency of pericytes in the central nervous system can cause increased permeability of the blood–brain barrier.

<span class="mw-page-title-main">Rostral migratory stream</span> One path neural stem cells take to reach the olfactory bulb


The rostral migratory stream (RMS) is a specialized migratory route found in the brain of some animals along which neuronal precursors that originated in the subventricular zone (SVZ) of the brain migrate to reach the main olfactory bulb (OB). The importance of the RMS lies in its ability to refine and even change an animal's sensitivity to smells, which explains its importance and larger size in the rodent brain as compared to the human brain, as our olfactory sense is not as developed. This pathway has been studied in the rodent, rabbit, and both the squirrel monkey and rhesus monkey. When the neurons reach the OB they differentiate into GABAergic interneurons as they are integrated into either the granule cell layer or periglomerular layer.

<span class="mw-page-title-main">Neuroimmune system</span>

The neuroimmune system is a system of structures and processes involving the biochemical and electrophysiological interactions between the nervous system and immune system which protect neurons from pathogens. It serves to protect neurons against disease by maintaining selectively permeable barriers, mediating neuroinflammation and wound healing in damaged neurons, and mobilizing host defenses against pathogens.

The biochemistry of Alzheimer's disease, the most common cause of dementia, is not yet very well understood. Alzheimer's disease (AD) has been identified as a proteopathy: a protein misfolding disease due to the accumulation of abnormally folded amyloid beta (Aβ) protein in the brain. Amyloid beta is a short peptide that is an abnormal proteolytic byproduct of the transmembrane protein amyloid-beta precursor protein (APP), whose function is unclear but thought to be involved in neuronal development. The presenilins are components of proteolytic complex involved in APP processing and degradation.

Glucose transporter 3, also known as solute carrier family 2, facilitated glucose transporter member 3 (SLC2A3) is a protein that in humans is encoded by the SLC2A3 gene. GLUT3 facilitates the transport of glucose across the plasma membranes of mammalian cells. GLUT3 is most known for its specific expression in neurons and has originally been designated as the neuronal GLUT. GLUT3 has been studied in other cell types with specific glucose requirements, including sperm, preimplantation embryos, circulating white blood cells and carcinoma cell lines.

<span class="mw-page-title-main">Subgranular zone</span>

The subgranular zone (SGZ) is a brain region in the hippocampus where adult neurogenesis occurs. The other major site of adult neurogenesis is the subventricular zone (SVZ) in the brain.

Gliotransmitters are chemicals released from glial cells that facilitate neuronal communication between neurons and other glial cells. They are usually induced from Ca2+ signaling, although recent research has questioned the role of Ca2+ in gliotransmitters and may require a revision of the relevance of gliotransmitters in neuronal signalling in general.

<span class="mw-page-title-main">Quinolinic acid</span> Dicarboxylic acid with pyridine backbone

Quinolinic acid, also known as pyridine-2,3-dicarboxylic acid, is a dicarboxylic acid with a pyridine backbone. It is a colorless solid. It is the biosynthetic precursor to niacin.

<span class="mw-page-title-main">Glymphatic system</span> System for waste clearance in the CNS of vertebrates

The glymphatic system is a system for waste clearance in the central nervous system (CNS) of vertebrates. According to this model, cerebrospinal fluid (CSF) flows into the paravascular space around cerebral arteries, combining with interstitial fluid (ISF) and parenchymal solutes, and exiting down venous paravascular spaces. The pathway consists of a para-arterial influx route for CSF to enter the brain parenchyma, coupled to a clearance mechanism for the removal of interstitial fluid (ISF) and extracellular solutes from the interstitial compartments of the brain and spinal cord. Exchange of solutes between CSF and ISF is driven primarily by arterial pulsation and regulated during sleep by the expansion and contraction of brain extracellular space. Clearance of soluble proteins, waste products, and excess extracellular fluid is accomplished through convective bulk flow of ISF, facilitated by astrocytic aquaporin 4 (AQP4) water channels.

Neuroinflammation is inflammation of the nervous tissue. It may be initiated in response to a variety of cues, including infection, traumatic brain injury, toxic metabolites, or autoimmunity. In the central nervous system (CNS), including the brain and spinal cord, microglia are the resident innate immune cells that are activated in response to these cues. The CNS is typically an immunologically privileged site because peripheral immune cells are generally blocked by the blood–brain barrier (BBB), a specialized structure composed of astrocytes and endothelial cells. However, circulating peripheral immune cells may surpass a compromised BBB and encounter neurons and glial cells expressing major histocompatibility complex molecules, perpetuating the immune response. Although the response is initiated to protect the central nervous system from the infectious agent, the effect may be toxic and widespread inflammation as well as further migration of leukocytes through the blood–brain barrier may occur.

Neuroangiogenesis is the coordinated growth of nerves and blood vessels. The nervous and blood vessel systems share guidance cues and cell-surface receptors allowing for this synchronised growth. The term neuroangiogenesis only came into use in 2002 and the process was previously known as neurovascular patterning. The combination of neurogenesis and angiogenesis is an essential part of embryonic development and early life. It is thought to have a role in pathologies such as endometriosis, brain tumors, and Alzheimer's disease.

<span class="mw-page-title-main">Pathophysiology of Parkinson's disease</span> Medical condition

The pathophysiology of Parkinson's disease is death of dopaminergic neurons as a result of changes in biological activity in the brain with respect to Parkinson's disease (PD). There are several proposed mechanisms for neuronal death in PD; however, not all of them are well understood. Five proposed major mechanisms for neuronal death in Parkinson's Disease include protein aggregation in Lewy bodies, disruption of autophagy, changes in cell metabolism or mitochondrial function, neuroinflammation, and blood–brain barrier (BBB) breakdown resulting in vascular leakiness.

<span class="mw-page-title-main">Functional ultrasound imaging</span> Ultrasound technique

Functional ultrasound imaging (fUS) is a medical ultrasound imaging technique of detecting or measuring changes in neural activities or metabolism, for example, the loci of brain activity, typically through measuring blood flow or hemodynamic changes. The method can be seen as an extension of Doppler imaging.

<span class="mw-page-title-main">Katerina Akassoglou</span> Greek neuroimmunologist

Katerina Akassoglou is a neuroimmunologist who is a Senior Investigator and Director of In Vivo Imaging Research at the Gladstone Institutes. Akassoglou holds faculty positions as a Professor of Neurology at the University of California, San Francisco. Akassoglou has pioneered investigations of blood-brain barrier integrity and development of neurological diseases. She found that compromised blood-brain barrier integrity leads to fibrinogen leakage into the brain inducing neurodegeneration. Akassoglou is internationally recognized for her scientific discoveries.

<span class="mw-page-title-main">Brain cell</span> Functional tissue of the brain

Brain cells make up the functional tissue of the brain. The rest of the brain tissue is structural or connective called the stroma which includes blood vessels. The two main types of cells in the brain are neurons, also known as nerve cells, and glial cells, also known as neuroglia.

VINE-seq is a method to isolate and molecularly characterize the vascular and perivascular cells of the human brain microvessels at single-nuclei resolution. This technique is achieved by combining various known laboratory-based strategies involving the mechanical dissociation of brain tissue samples into single cells, density gradient centrifugation and filtration to isolate nuclei of microvessels, fluorescence-activated cell sorting (FACs) of cellular populations and droplet-based single-nuclei RNA sequencing (drop-snRNA-seq). Altogether, this generates a single-nuclei transcriptomic profile of the various cell types present in the vasculature of the brain. Through processing and analyzing the single-nuclei transcriptomic data, the heterogeneity within and between cell types can be distinguished to construct the molecular landscape of the human brain vasculature that was not previously done before.

Nozomi Nishimura is an American biomedical engineer who is an associate professor at Cornell University. She was awarded the L'Oréal for Women in Science Fellowship in 2009 and was inducted into the 2024 Class of the AIMBE College of Fellows for her research in intravital microscopy contributing to the understanding of microscale physiology.

References

  1. 1 2 3 4 5 6 7 8 9 10 11 12 Iadecola C (September 2017). "The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease". Neuron. 96 (1): 17–42. doi:10.1016/j.neuron.2017.07.030. PMC   5657612 . PMID   28957666.
  2. 1 2 Stackhouse TL, Mishra A (2021-07-12). "Neurovascular Coupling in Development and Disease: Focus on Astrocytes". Frontiers in Cell and Developmental Biology. 9: 702832. doi: 10.3389/fcell.2021.702832 . PMC   8313501 . PMID   34327206.
  3. 1 2 3 4 Muoio V, Persson PB, Sendeski MM (April 2014). "The neurovascular unit - concept review". Acta Physiologica. 210 (4): 790–798. doi: 10.1111/apha.12250 . PMID   24629161. S2CID   25274791.
  4. Cauli B, Tong XK, Rancillac A, Serluca N, Lambolez B, Rossier J, et al. (October 2004). "Cortical GABA interneurons in neurovascular coupling: relays for subcortical vasoactive pathways". The Journal of Neuroscience. 24 (41): 8940–8949. doi:10.1523/JNEUROSCI.3065-04.2004. PMC   6730057 . PMID   15483113. The expression of vasoactive intestinal peptide (VIP) or nitric oxide synthase (NOS) in interneurons induces dilation, while somatostatin (SOM) induces contraction. Direct perfusion of VIP and NO donors onto the slices caused microvessel dilation, whereas neuropeptide Y (NPY) and SOM induced vasoconstriction. Vasomotor interneurons established contacts with local microvessels and received somatic and dendritic afferents from acetylcholine (ACh) and serotonin (5-HT) pathways, varying by interneuron subtype. Our results demonstrate the capability of specific subsets of cortical GABA interneurons to transmute neuronal signals into vascular responses and suggest that they could serve as local integrators of neurovascular coupling for subcortical vasoactive pathways.
  5. Carmignoto G, Gómez-Gonzalo M (May 2010). "The contribution of astrocyte signalling to neurovascular coupling". Brain Research Reviews. 63 (1–2): 138–148. doi:10.1016/j.brainresrev.2009.11.007. PMID   19948187.
  6. Rancillac A, Rossier J, Guille M, Tong XK, Geoffroy H, Amatore C, et al. (June 2006). "Glutamatergic Control of Microvascular Tone by Distinct GABA Neurons in the Cerebellum". The Journal of Neuroscience. 26 (26): 6997–7006. doi:10.1523/JNEUROSCI.5515-05.2006. PMC   6673912 . PMID   16807329. Cerebellar stellate and Purkinje cells play distinct roles in neurovascular coupling by dilating and constricting neighboring microvessels, respectively. This highlights the specialized functions of different neuron types in regulating cerebral blood flow.
  7. Filosa JA (2010). "Vascular tone and neurovascular coupling: considerations toward an improved in vitro model". Frontiers in Neuroenergetics. 2. doi: 10.3389/fnene.2010.00016 . PMC   2928708 . PMID   20802803.
  8. 1 2 3 Phillips AA, Chan FH, Zheng MM, Krassioukov AV, Ainslie PN (April 2016). "Neurovascular coupling in humans: Physiology, methodological advances and clinical implications". Journal of Cerebral Blood Flow and Metabolism. 36 (4): 647–664. doi:10.1177/0271678X15617954. PMC   4821024 . PMID   26661243.
  9. Cipolla MJ (2009). "Chapter 5, Control of Cerebral Blood Flow". The Cerebral Circulation. San Rafael (CA): Morgan & Claypool Life Sciences.
  10. Rancillac A, Rossier J, Guille M, Tong XK, Geoffroy H, Amatore C, et al. (June 2006). "Glutamatergic Control of Microvascular Tone by Distinct GABA Neurons in the Cerebellum". The Journal of Neuroscience. 26 (26): 6997–7006. doi:10.1523/JNEUROSCI.5515-05.2006. PMC   6673912 . PMID   16807329.
  11. 1 2 Ahmad A, Patel V, Xiao J, Khan MM (November 2020). "The Role of Neurovascular System in Neurodegenerative Diseases". Molecular Neurobiology. 57 (11): 4373–4393. doi:10.1007/s12035-020-02023-z. PMID   32725516. S2CID   220843844.
  12. 1 2 Daneman R, Prat A (January 2015). "The blood-brain barrier". Cold Spring Harbor Perspectives in Biology. 7 (1): a020412. doi:10.1101/cshperspect.a020412. PMC   4292164 . PMID   25561720.
  13. Pasley BN, Freeman RD (2008-03-11). "Neurovascular coupling". Scholarpedia. 3 (3): 5340. Bibcode:2008SchpJ...3.5340P. doi: 10.4249/scholarpedia.5340 . ISSN   1941-6016.
  14. Rancillac A, Rossier J, Guille M, Tong XK, Geoffroy H, Amatore C, et al. (June 2006). "Glutamatergic Control of Microvascular Tone by Distinct GABA Neurons in the Cerebellum". The Journal of Neuroscience. 26 (26): 6997–7006. doi:10.1523/JNEUROSCI.5515-05.2006. PMC   6673912 . PMID   16807329.
  15. Cauli B, Tong XK, Rancillac A, Serluca N, Lambolez B, Rossier J, et al. (October 2004). "Cortical GABA interneurons in neurovascular coupling: relays for subcortical vasoactive pathways". The Journal of Neuroscience. 24 (41): 8940–8949. doi:10.1523/JNEUROSCI.3065-04.2004. PMC   6730057 . PMID   15483113.
  16. Scharbarg E, Daenens M, Lemaître F, Geoffroy H, Guille-Collignon M, Gallopin T, et al. (January 2016). "Astrocyte-derived adenosine is central to the hypnogenic effect of glucose". Scientific Reports. 6 (1): 19107. Bibcode:2016NatSR...619107S. doi:10.1038/srep19107. PMC   4709579 . PMID   26755200. Using infrared videomicroscopy on ex vivo brain slices, we established that glucose induces vasodilation specifically in the Ventrolateral Preoptic Nucleus (VLPO) via astrocytic release of adenosine.
  17. Scharbarg E, Walter A, Lecoin L, Gallopin T, Lemaître F, Guille-Collignon M, et al. (March 2023). "Prostaglandin D2 Controls Local Blood Flow and Sleep-Promoting Neurons in the VLPO via Astrocyte-Derived Adenosine" (PDF). ACS Chemical Neuroscience. 14 (6): 1063–1070. doi:10.1021/acschemneuro.2c00660. PMID   36847485. Measurements of vasodilatory responses and electrophysiological recordings reveal that, in response to PGD2 application, adenosine release induces A2A receptor (A2AR)-mediated dilation of blood vessels and activation of Ventrolateral Preoptic Nucleus (VLPO) sleep-promoting neurons. Collectively, our results elucidate the PGD2 signaling pathway in the VLPO, demonstrating its role in controlling local blood flow and activating sleep-promoting neurons via astrocyte-derived adenosine.
  18. Chen S, Wang Z, Zhang D, Wang A, Chen L, Cheng H, et al. (October 2020). "Miniature Fluorescence Microscopy for Imaging Brain Activity in Freely-Behaving Animals". Neuroscience Bulletin. 36 (10): 1182–1190. doi:10.1007/s12264-020-00561-z. PMC   7532237 . PMID   32797396.
  19. Mai-Morente SP, Marset VM, Blanco F, Isasi EE, Abudara V (May 2021). "A nuclear fluorescent dye identifies pericytes at the neurovascular unit". Journal of Neurochemistry. 157 (4): 1377–1391. doi:10.1111/jnc.15193. hdl: 20.500.12008/26846 . PMID   32974913.
  20. 1 2 Sanderson MJ, Smith I, Parker I, Bootman MD (October 2014). "Fluorescence microscopy". Cold Spring Harbor Protocols. 2014 (10): pdb.top071795. doi:10.1101/pdb.top071795. PMC   4711767 . PMID   25275114.
  21. Nahirney PC, Tremblay ME (2021). "Brain Ultrastructure: Putting the Pieces Together". Frontiers in Cell and Developmental Biology. 9: 629503. doi: 10.3389/fcell.2021.629503 . PMC   7930431 . PMID   33681208.
  22. Knott G, Marchman H, Wall D, Lich B (March 2008). "Serial section scanning electron microscopy of adult brain tissue using focused ion beam milling". The Journal of Neuroscience. 28 (12): 2959–2964. doi:10.1523/JNEUROSCI.3189-07.2008. PMC   6670719 . PMID   18353998.
  23. Kubota Y, Sohn J, Kawaguchi Y (2018). "Large Volume Electron Microscopy and Neural Microcircuit Analysis". Frontiers in Neural Circuits. 12: 98. doi: 10.3389/fncir.2018.00098 . PMC   6240581 . PMID   30483066.
  24. "Magnetic Resonance Imaging (MRI)". National Institute of Biomedical Imaging and Bioengineering. Retrieved 2023-06-19.
  25. Mueller BA, Lim KO, Hemmy L, Camchong J (September 2015). "Diffusion MRI and its Role in Neuropsychology". Neuropsychology Review. 25 (3): 250–271. doi:10.1007/s11065-015-9291-z. PMC   4807614 . PMID   26255305.
  26. 1 2 Fujimoto JG, Pitris C, Boppart SA, Brezinski ME (2000-01-01). "Optical coherence tomography: an emerging technology for biomedical imaging and optical biopsy". Neoplasia. 2 (1–2): 9–25. doi:10.1038/sj.neo.7900071. PMC   1531864 . PMID   10933065.
  27. 1 2 Anagnostakou V, Ughi GJ, Puri AS, Gounis MJ (October 2021). "Optical Coherence Tomography for Neurovascular Disorders". Neuroscience. Brain imaging. 474: 134–144. doi:10.1016/j.neuroscience.2021.06.008. PMID   34126186.
  28. 1 2 "Cerebrovascular Disease". Penn Medicine. Philadelphia, PA: University of Pennsylvania. Retrieved 2023-06-19.
  29. 1 2 3 4 5 Stanimirovic DB, Friedman A (July 2012). "Pathophysiology of the neurovascular unit: disease cause or consequence?". Journal of Cerebral Blood Flow and Metabolism. 32 (7): 1207–1221. doi:10.1038/jcbfm.2012.25. PMC   3390807 . PMID   22395208.
  30. 1 2 3 Shabir O, Berwick J, Francis SE (October 2018). "Neurovascular dysfunction in vascular dementia, Alzheimer's and atherosclerosis". BMC Neuroscience. 19 (1): 62. doi: 10.1186/s12868-018-0465-5 . PMC   6192291 . PMID   30333009.
  31. 1 2 Nelson AR, Sweeney MD, Sagare AP, Zlokovic BV (May 2016). "Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease". Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. Vascular contributions to cognitive impairment and dementia (VCID). 1862 (5): 887–900. doi:10.1016/j.bbadis.2015.12.016. PMC   4821735 . PMID   26705676.
  32. 1 2 Iadecola C (May 2004). "Neurovascular regulation in the normal brain and in Alzheimer's disease". Nature Reviews. Neuroscience. 5 (5): 347–360. doi:10.1038/nrn1387. PMID   15100718. S2CID   36555564.
  33. 1 2 Lin CY, Hsu YH, Lin MH, Yang TH, Chen HM, Chen YC, et al. (December 2013). "Neurovascular abnormalities in humans and mice with Huntington's disease". Experimental Neurology. 250: 20–30. doi:10.1016/j.expneurol.2013.08.019. PMID   24036415. S2CID   9613243.
  34. Kapasi A, Schneider JA (May 2016). "Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons". Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1862 (5): 878–886. doi: 10.1016/j.bbadis.2015.12.023 . PMC   11062590 . PMID   26769363.
  35. Kumar A, Sidhu J, Goyal A, Tsao JW (2023). "Alzheimer Disease". StatPearls. Treasure Island (FL): StatPearls Publishing. PMID   29763097 . Retrieved 2023-06-19.
  36. Soto-Rojas LO, Pacheco-Herrero M, Martínez-Gómez PA, Campa-Córdoba BB, Apátiga-Pérez R, Villegas-Rojas MM, et al. (February 2021). "The Neurovascular Unit Dysfunction in Alzheimer's Disease". International Journal of Molecular Sciences. 22 (4): 2022. doi: 10.3390/ijms22042022 . PMC   7922832 . PMID   33670754.
  37. 1 2 3 Rius-Pérez S, Tormos AM, Pérez S, Taléns-Visconti R (March 2018). "Vascular pathology: Cause or effect in Alzheimer disease?". Neurologia. 33 (2): 112–120. doi: 10.1016/j.nrleng.2015.07.008 . PMID   26385017.
  38. Kalaria RN, Ballard C (1999). "Overlap between pathology of Alzheimer disease and vascular dementia". Alzheimer Disease and Associated Disorders. 13 (Suppl 3): S115–S123. doi:10.1097/00002093-199912003-00017. PMID   10609690.
  39. 1 2 Beishon L, Panerai RB (2021). "The Neurovascular Unit in Dementia: An Opinion on Current Research and Future Directions". Frontiers in Aging Neuroscience. 13: 721937. doi: 10.3389/fnagi.2021.721937 . PMC   8355558 . PMID   34393765.
  40. Nopoulos PC (March 2016). "Huntington disease: a single-gene degenerative disorder of the striatum". Dialogues in Clinical Neuroscience. 18 (1): 91–98. doi:10.31887/DCNS.2016.18.1/pnopoulos. PMC   4826775 . PMID   27069383.
  41. Novak MJ, Tabrizi SJ (June 2010). "Huntington's disease". BMJ. 340: c3109. doi:10.1136/bmj.c3109. PMID   20591965. S2CID   16061768.
  42. Jakel RJ, Maragos WF (June 2000). "Neuronal cell death in Huntington's disease: a potential role for dopamine". Trends in Neurosciences. 23 (6): 239–245. doi:10.1016/s0166-2236(00)01568-x. PMID   10838590. S2CID   25754639.
  43. Chen JJ, Salat DH, Rosas HD (January 2012). "Complex relationships between cerebral blood flow and brain atrophy in early Huntington's disease". NeuroImage. 59 (2): 1043–1051. doi:10.1016/j.neuroimage.2011.08.112. PMC   3787075 . PMID   21945790.
  44. Hsiao HY, Chen YC, Huang CH, Chen CC, Hsu YH, Chen HM, et al. (August 2015). "Aberrant astrocytes impair vascular reactivity in Huntington disease". Annals of Neurology. 78 (2): 178–192. doi:10.1002/ana.24428. PMID   25914140. S2CID   30714012.
  45. Drouin-Ouellet J, Sawiak SJ, Cisbani G, Lagacé M, Kuan WL, Saint-Pierre M, et al. (August 2015). "Cerebrovascular and blood-brain barrier impairments in Huntington's disease: Potential implications for its pathophysiology". Annals of Neurology. 78 (2): 160–177. doi:10.1002/ana.24406. PMID   25866151. S2CID   15993646.
  46. Chan ST, Mercaldo ND, Kwong KK, Hersch SM, Rosas HD (2021). "Impaired Cerebrovascular Reactivity in Huntington's Disease". Frontiers in Physiology. 12: 663898. doi: 10.3389/fphys.2021.663898 . PMC   8334185 . PMID   34366879.
  47. Garcia FJ, Sun N, Lee H, Godlewski B, Mathys H, Galani K, et al. (March 2022). "Single-cell dissection of the human brain vasculature". Nature. 603 (7903): 893–899. Bibcode:2022Natur.603..893G. doi:10.1038/s41586-022-04521-7. PMC   9680899 . PMID   35158371.