87 results on '"Jeffrey J. Iliff"'
Search Results
2. Interrogation of dynamic glucose-enhanced MRI and fluorescence-based imaging reveals a perturbed glymphatic network in Huntington’s disease
- Author
-
Hongshuai Liu, Lin Chen, Chuangchuang Zhang, Chang Liu, Yuguo Li, Liam Cheng, Zhiliang Wei, Ziqin Zhang, Hanzhang Lu, Peter C. M. van Zijl, Jeffrey J. Iliff, Jiadi Xu, and Wenzhen Duan
- Subjects
Article - Abstract
Huntington’s disease (HD) is a neurodegenerative disorder that presents with progressive motor, mental, and cognitive impairment leading to early disability and mortality. The accumulation of mutant huntingtin protein aggregates in neurons is a pathological hallmark of HD. The glymphatic system, a brain-wide perivascular network, facilitates the exchange of interstitial fluid (ISF) and cerebrospinal fluid (CSF), supporting interstitial solute clearance including abnormal proteins from mammalian brains. In this study, we employed dynamic glucose-enhanced (DGE) MRI to measure D-glucose clearance from CSF as a tool to assess CSF clearance capacity to predict glymphatic function in a mouse model of HD. Our results demonstrate significantly diminished CSF clearance efficiency in premanifest zQ175 HD mice. The impairment of CSF clearance of D-glucose, measured by DGE MRI, worsened with disease progression. These DGE MRI findings in compromised glymphatic function in HD mice were further confirmed with fluorescence-based imaging of glymphatic CSF tracer influx, suggesting an impaired glymphatic function in premanifest stage of HD. Moreover, expression of the astroglial water channel aquaporin-4 (AQP4) in the perivascular compartment, a key mediator of glymphatic function, was significantly diminished in both HD mouse brain as well as postmortem human HD brain. Our data, acquired using a clinically translatable MRI approach, indicate a perturbed glymphatic network in the HD brain as early as in the premanifest stage. Further validation of these findings in clinical studies should provide insights into potential of glymphatic clearance as a HD biomarker and for glymphatic functioning as a disease-modifying therapeutic target for HD.
- Published
- 2023
- Full Text
- View/download PDF
3. The Bidirectional Link Between Sleep Disturbances and Traumatic Brain Injury Symptoms: A Role for Glymphatic Dysfunction?
- Author
-
Jeffrey J. Iliff, Miranda M. Lim, and Juan Piantino
- Subjects
Sleep Wake Disorders ,medicine.medical_specialty ,business.industry ,Traumatic brain injury ,medicine.disease ,Sleep in non-human animals ,Article ,Poor sleep ,Therapeutic approach ,Physical medicine and rehabilitation ,Subjective sleep ,Brain Injuries, Traumatic ,Concussion ,Humans ,Medicine ,Glymphatic system ,Sleep ,business ,Cognitive impairment ,Glymphatic System ,Brain Concussion ,Biological Psychiatry - Abstract
Mild traumatic brain injury (mTBI), often referred to as concussion, is a major cause of morbidity and mortality worldwide. Sleep disturbances are common after mTBI. Moreover, subjects who develop subjective sleep complaints after mTBI also report more severe somatic, mental health, and cognitive impairment and take longer to recover from mTBI sequelae. Despite many previous studies addressing the role of sleep in post-mTBI morbidity, the mechanisms linking sleep to recovery after mTBI remain poorly understood. The glymphatic system is a brain-wide network that supports fluid movement through the cerebral parenchyma and the clearance of interstitial solutes and wastes from the brain. Notably, the glymphatic system is active primarily during sleep. Clearance of cellular byproducts related to somatic, mental health, and neurodegenerative processes (e.g., amyloid-β and tau, among others) depends in part on intact glymphatic function, which becomes impaired after mTBI. In this viewpoint, we review the current knowledge regarding the association between sleep disturbances and post-mTBI symptoms. We also discuss the role of glymphatic dysfunction as a potential link between mTBI, sleep disruption, and post-traumatic morbidity. We outline a model where glymphatic dysfunction and sleep disruption caused by mTBI may have an additive effect on waste clearance, leading to cerebral dysfunction and impaired recovery. Finally, we review the novel techniques being developed to examine glymphatic function in humans and explore potential interventions to alter glymphatic exchange that may offer a novel therapeutic approach to those suffering from poor sleep and prolonged symptoms after mTBI.
- Published
- 2022
- Full Text
- View/download PDF
4. Author response for 'Quantification Approaches for Magnetic Resonance Imaging Following Intravenous Gadolinium Injection: A Window into Brain‐Wide Glymphatic Function'
- Author
-
null Sutton B. Richmond, null Swati Rane, null Moriah R. Hanson, null Mehmet Albayram, null Jeffrey J. Iliff, null Dawn Kernagis, null Jens T. Rosenberg, and null Rachael D. Seidler
- Published
- 2023
- Full Text
- View/download PDF
5. Mapping the lymphatic system across body scales and expertise domains : A report from the 2021 National Heart, Lung, and Blood Institute workshop at the Boston Lymphatic Symposium
- Author
-
Dhruv Singhal, Katy Börner, Elliot L. Chaikof, Michael Detmar, Maija Hollmén, Jeffrey J. Iliff, Maxim Itkin, Taija Makinen, Guillermo Oliver, Timothy P. Padera, Ellen M. Quardokus, Andrea J. Radtke, Hiroo Suami, Griffin M. Weber, Ilsa I. Rovira, Selen C. Muratoglu, and Zorina S. Galis
- Subjects
Fysiologi ,Physiology ,Lymphatic disease ,Lymphedema ,Lymphatic research ,Mapping ,Lymphatic anatomy ,Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) ,lymphedema ,lymphatic anatomy ,Physiology (medical) ,mapping ,Medicinsk bioteknologi (med inriktning mot cellbiologi (inklusive stamcellsbiologi), molekylärbiologi, mikrobiologi, biokemi eller biofarmaci) ,lymphatic disease ,lymphatic research - Abstract
Enhancing our understanding of lymphatic anatomy from the microscopic to the anatomical scale is essential to discern how the structure and function of the lymphatic system interacts with different tissues and organs within the body and contributes to health and disease. The knowledge of molecular aspects of the lymphatic network is fundamental to understand the mechanisms of disease progression and prevention. Recent advances in mapping components of the lymphatic system using state of the art single cell technologies, the identification of novel biomarkers, new clinical imaging efforts, and computational tools which attempt to identify connections between these diverse technologies hold the potential to catalyze new strategies to address lymphatic diseases such as lymphedema and lipedema. This manuscript summarizes current knowledge of the lymphatic system and identifies prevailing challenges and opportunities to advance the field of lymphatic research as discussed by the experts in the workshop., Frontiers in Physiology, 14, ISSN:1664-042X
- Published
- 2023
6. The glymphatic system: Current understanding and modeling
- Author
-
Tomas Bohr, Poul G. Hjorth, Sebastian C. Holst, Sabina Hrabětová, Vesa Kiviniemi, Tuomas Lilius, Iben Lundgaard, Kent-Andre Mardal, Erik A. Martens, Yuki Mori, U. Valentin Nägerl, Charles Nicholson, Allen Tannenbaum, John H. Thomas, Jeffrey Tithof, Helene Benveniste, Jeffrey J. Iliff, Douglas H. Kelley, and Maiken Nedergaard
- Subjects
Neuroanatomy ,Multidisciplinary ,Systems biology ,Neuroscience - Abstract
We review theoretical and numerical models of the glymphatic system, which circulates cerebrospinal fluid and interstitial fluid around the brain, facilitating solute transport. Models enable hypothesis development and predictions of transport, with clinical applications including drug delivery, stroke, cardiac arrest, and neurodegenerative disorders like Alzheimer's disease. We sort existing models into broad categories by anatomical function: Perivascular flow, transport in brain parenchyma, interfaces to perivascular spaces, efflux routes, and links to neuronal activity. Needs and opportunities for future work are highlighted wherever possible; new models, expanded models, and novel experiments to inform models could all have tremendous value for advancing the field.
- Published
- 2022
- Full Text
- View/download PDF
7. The effect of aquaporin-4 mis-localization on Aβ deposition in mice
- Author
-
Taylor J. Pedersen, Samantha A. Keil, Warren Han, Marie X. Wang, and Jeffrey J. Iliff
- Subjects
Neurology - Published
- 2023
- Full Text
- View/download PDF
8. Longitudinal MRI-visible perivascular space (PVS) changes with long-duration spaceflight
- Author
-
Kathleen E. Hupfeld, Sutton B. Richmond, Heather R. McGregor, Daniel L. Schwartz, Madison N. Luther, Nichole E. Beltran, Igor S. Kofman, Yiri E. De Dios, Roy F. Riascos, Scott J. Wood, Jacob J. Bloomberg, Ajitkumar P. Mulavara, Lisa C. Silbert, Jeffrey J. Iliff, Rachael D. Seidler, and Juan Piantino
- Subjects
Multidisciplinary - Abstract
Humans are exposed to extreme environmental stressors during spaceflight and return with alterations in brain structure and shifts in intracranial fluids. To date, no studies have evaluated the effects of spaceflight on perivascular spaces (PVSs) within the brain, which are believed to facilitate fluid drainage and brain homeostasis. Here, we examined how the number and morphology of magnetic resonance imaging (MRI)-visible PVSs are affected by spaceflight, including prior spaceflight experience. Fifteen astronauts underwent six T1-weighted 3 T MRI scans, twice prior to launch and four times following their return to Earth after ~ 6-month missions to the International Space Station. White matter MRI-visible PVS number and morphology were calculated using an established, automated segmentation algorithm. We validated our automated segmentation algorithm by comparing algorithm PVS counts with those identified by two trained raters in 50 randomly selected slices from this cohort; the automated algorithm performed similarly to visual ratings (r(48) = 0.77, p k) > 0.50). Among the astronaut cohort, we found that novice astronauts showed an increase in total PVS volume from pre- to post-flight, whereas experienced crewmembers did not (p = 0.020), suggesting that experienced astronauts may exhibit holdover effects from prior spaceflight(s). Greater pre-flight PVS load was associated with more prior flight experience (r = 0.60–0.71), though these relationships did not reach statistical significance (p > 0.05). Pre- to post-flight changes in ventricular volume were not significantly associated with changes in PVS characteristics, and the presence of spaceflight associated neuro-ocular syndrome (SANS) was not associated with PVS number or morphology. Together, these findings demonstrate that PVSs can be consistently identified on T1-weighted MRI scans, and that spaceflight is associated with PVS changes. Specifically, prior spaceflight experience may be an important factor in determining PVS characteristics.
- Published
- 2022
- Full Text
- View/download PDF
9. When the air hits your brain: decreased arterial pulsatility after craniectomy leading to impaired glymphatic flow
- Author
-
Benjamin A. Plog, Nanhong Lou, Clifford A. Pierre, Alex Cove, H. Mark Kenney, Emi Hitomi, Hongyi Kang, Jeffrey J. Iliff, Douglas M. Zeppenfeld, Maiken Nedergaard, and G. Edward Vates
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Microglia ,business.industry ,medicine.medical_treatment ,General Medicine ,Cranioplasty ,Article ,03 medical and health sciences ,Skull ,0302 clinical medicine ,medicine.anatomical_structure ,Gliosis ,Interstitial fluid ,Parenchyma ,Cranial cavity ,medicine ,Glymphatic system ,medicine.symptom ,business ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
OBJECTIVECranial neurosurgical procedures can cause changes in brain function. There are many potential explanations, but the effect of simply opening the skull has not been addressed, except for research into syndrome of the trephined. The glymphatic circulation, by which CSF and interstitial fluid circulate through periarterial spaces, brain parenchyma, and perivenous spaces, depends on arterial pulsations to provide the driving force for bulk flow; opening the cranial cavity could dampen this force. The authors hypothesized that a craniectomy, without any other pathological insult, is sufficient to alter brain function due to reduced arterial pulsatility and decreased glymphatic flow. Furthermore, they postulated that glymphatic impairment would produce activation of astrocytes and microglia; with the reestablishment of a closed cranial compartment, the glymphatic impairment, astrocytic/microglial activation, and neurobehavioral decline caused by opening the cranial compartment might be reversed.METHODSUsing two-photon in vivo microscopy, the pulsatility index of cortical vessels was quantified through a thinned murine skull and then again after craniectomy. Glymphatic influx was determined with ex vivo fluorescence microscopy of mice 0, 14, 28, and 56 days following craniectomy or cranioplasty; brain sections were immunohistochemically labeled for GFAP and CD68. Motor and cognitive performance was quantified with rotarod and novel object recognition tests at baseline and 14, 21, and 28 days following craniectomy or cranioplasty.RESULTSPenetrating arterial pulsatility decreased significantly and bilaterally following unilateral craniectomy, producing immediate and chronic impairment of glymphatic CSF influx in the ipsilateral and contralateral brain parenchyma. Craniectomy-related glymphatic dysfunction was associated with an astrocytic and microglial inflammatory response, as well as with the development of motor and cognitive deficits. Recovery of glymphatic flow preceded reduced gliosis and return of normal neurological function, and cranioplasty accelerated this recovery.CONCLUSIONSCraniectomy causes glymphatic dysfunction, gliosis, and changes in neurological function in this murine model of syndrome of the trephined.
- Published
- 2020
- Full Text
- View/download PDF
10. Coniferaldehyde attenuates Alzheimer's pathology via activation of Nrf2 and its targets
- Author
-
Danfeng Zheng, Xiaoda Yang, Tessandra Stewart, Taotao Wei, Xue Li, Yaqiong Dong, Lan Yuan, Li-dan Bai, Min Shi, Jing Zhang, Jeffrey J. Iliff, and Ting Xu
- Subjects
0301 basic medicine ,Agonist ,Male ,Pathology ,medicine.medical_specialty ,medicine.drug_class ,NF-E2-Related Factor 2 ,Medicine (miscellaneous) ,Hippocampus ,Mice, Transgenic ,Neuroprotection ,Nrf2 ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Acrolein ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Neurons ,Amyloid beta-Peptides ,business.industry ,Brain ,coniferaldehyde ,Extracellular vesicle ,Alzheimer's disease ,Aβ clearance ,In vitro ,3. Good health ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Neuroprotective Agents ,Cerebral cortex ,neuroprotection ,Female ,Neuron ,business ,030217 neurology & neurosurgery ,Research Paper - Abstract
Background: Alzheimer's disease (AD) currently lacks a cure. Because substantial neuronal damage usually occurs before AD is advanced enough for diagnosis, the best hope for disease-modifying AD therapies likely relies on early intervention or even prevention, and targeting multiple pathways implicated in early AD pathogenesis rather than focusing exclusively on excessive production of β-amyloid (Aβ) species. Methods: Coniferaldehyde (CFA), a food flavoring and agonist of NF-E2-related factor 2 (Nrf2), was selected by multimodal in vitro screening, followed by investigation of several downstream effects potentially involved. Furthermore, in the APP/PS1 AD mouse model, the therapeutic effects of CFA (0.2 mmol kg-1d-1) were tested beginning at 3 months of age. Behavioral phenotypes related to learning and memory capacity, brain pathology and biochemistry, including Aβ transport, were assessed at different time intervals. Results: CFA promoted neuron viability and showed potent neuroprotective effects, especially on mitochondrial structure and functions. In addition, CFA greatly enhanced the brain clearance of Aβ in both free and extracellular vesicle (EV)-contained Aβ forms. In the APP/PS1 mouse model, CFA effectively abolished brain Aβ deposits and reduced the level of toxic soluble Aβ peptides, thus eliminating AD-like pathological changes in the hippocampus and cerebral cortex and preserving learning and memory capacity of the mice. Conclusion: The experimental evidence overall indicated that Nrf2 activation may contribute to the potent anti-AD effects of CFA. With an excellent safety profile, further clinical investigation of coniferaldehyde might bring hope for AD prevention/therapy.
- Published
- 2020
11. Impaired Glymphatic Function and Pulsation Alterations in a Mouse Model of Vascular Cognitive Impairment
- Author
-
Mosi Li, Akihiro Kitamura, Joshua Beverley, Juraj Koudelka, Jessica Duncombe, Ross Lennen, Maurits A. Jansen, Ian Marshall, Bettina Platt, Ulrich K. Wiegand, Roxana O. Carare, Rajesh N. Kalaria, Jeffrey J. Iliff, and Karen Horsburgh
- Subjects
vascular pulsation ,Aging ,cerebral amyloid angiopathy (CAA) ,Cognitive Neuroscience ,amyloid-β (Aβ) ,carotid stenosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,glymphatic function ,vascular cognitive impairment ,RC321-571 ,Neuroscience ,Original Research - Abstract
Large vessel disease and carotid stenosis are key mechanisms contributing to vascular cognitive impairment (VCI) and dementia. Our previous work, and that of others, using rodent models, demonstrated that bilateral common carotid stenosis (BCAS) leads to cognitive impairment via gradual deterioration of the neuro-glial-vascular unit and accumulation of amyloid-β (Aβ) protein. Since brain-wide drainage pathways (glymphatic) for waste clearance, including Aβ removal, have been implicated in the pathophysiology of VCI via glial mechanisms, we hypothesized that glymphatic function would be impaired in a BCAS model and exacerbated in the presence of Aβ. Male wild-type and Tg-SwDI (model of microvascular amyloid) mice were subjected to BCAS or sham surgery which led to a reduction in cerebral perfusion and impaired spatial learning acquisition and cognitive flexibility. After 3 months survival, glymphatic function was evaluated by cerebrospinal fluid (CSF) fluorescent tracer influx. We demonstrated that BCAS caused a marked regional reduction of CSF tracer influx in the dorsolateral cortex and CA1-DG molecular layer. In parallel to these changes increased reactive astrogliosis was observed post-BCAS. To further investigate the mechanisms that may lead to these changes, we measured the pulsation of cortical vessels. BCAS impaired vascular pulsation in pial arteries in WT and Tg-SwDI mice. Our findings show that BCAS influences VCI and that this is paralleled by impaired glymphatic drainage and reduced vascular pulsation. We propose that these additional targets need to be considered when treating VCI.
- Published
- 2022
- Full Text
- View/download PDF
12. Sleep Dependent Changes of Lactate Concentration in Human Brain
- Author
-
Selda Yildiz, Miranda M. Lim, Manoj K. Sammi, Katherine Powers, Charles F. Murchison, Jeffrey J. Iliff, and William D. Rooney
- Abstract
Lactate is an important cellular metabolite that is present at high concentrations in the brain, both within cells and in the extracellular space between cells. Small animal studies demonstrated high extracellular concentrations of lactate during wakefulness with reductions during sleep and/or anesthesia with a recent study suggesting the glymphatic activity as the mechanism for the reduction of lactate concentrations. We have recently developed a rigorous non-invasive imaging approach combining simultaneous magnetic resonance spectroscopy (MRS) and polysomnography (PSG) measurements, and here, we present the first in-vivo evaluation of brain lactate levels during sleep-wake cycles in young healthy humans. First, we collected single voxel proton MRS (1H-MRS) data at the posterior cingulate with high temporal resolution (every 7.5 sec), and simultaneously recorded PSG data while temporally registering with 1H-MRS time-series. Second, we evaluated PSG data in 30 s epochs, and classified into four stages Wake (W), Non-REM sleep stage 1 (N1), Non-REM sleep stage 2 (N2), and Non-REM sleep stage 3 (N3). Third, we determined lactate signal intensity from each 7.5-s spectrum, normalized to corresponding water signal, and averaged over 30-s for each PSG epoch. In examinations of nine healthy participants (four females, five males; mean age 24.2 (±2; SD) years; age range: 21-27 years) undergoing up to 3-hour simultaneous MRS/PSG recordings, we observed a group mean reduction of [4.9 ± 4.9] % in N1, [10.4 ± 5.2] % in N2, and [24.0 ± 5.8] % in N3 when compared to W. Our finding is consistent with more than 70 years of invasive lactate measurements from small animal studies. In addition, reduced brain lactate was accompanied by a significant reduction the apparent diffusion coefficient of brain lactate. Taken together, these findings are consistent with the loss of lactate from the extracellular space during sleep while suggesting lactate metabolism is altered and/or lactate clearance via glymphatic exchange is increased during sleep.Significance StatementThis study describes a non-invasive magnetic resonance spectroscopy/polysomnography technique that allows rigorous measurement of brain metabolite levels together with simultaneous characterization of brain arousal state as either wakeful or one of the several sleep states. The results provide the first in-vivo demonstration of reductions in brain lactate concentration and diffusivity during sleep versus wakefulness in young healthy human brain. These findings are consistent with invasive small-animal studies showing the loss of extracellular lactate during sleep, and support the notion of altered lactate metabolism and/or increased glymphatic activity in sleeping human brain.
- Published
- 2021
- Full Text
- View/download PDF
13. Quantitative analysis of macroscopic solute transport in the murine brain
- Author
-
Martin M. Pike, Lori Ray, Matthew J. Simon, Jeffrey J. Iliff, and Jeffrey J. Heys
- Subjects
Male ,Perivascular transport ,Brain transport ,Interstitial transport ,Convection ,Biotransport ,Mice ,Cellular and Molecular Neuroscience ,Murine brain ,Glymphatic ,Developmental Neuroscience ,Animals ,RC346-429 ,Chemistry ,Research ,Brain ,Biological Transport ,General Medicine ,Models, Theoretical ,Magnetic Resonance Imaging ,Mice, Inbred C57BL ,Dynamic contrast-enhanced MRI ,Neurology ,Biophysics ,Female ,Neurology. Diseases of the nervous system ,Quantitative analysis (chemistry) ,Glymphatic System - Abstract
Background Understanding molecular transport in the brain is critical to care and prevention of neurological disease and injury. A key question is whether transport occurs primarily by diffusion, or also by convection or dispersion. Dynamic contrast-enhanced (DCE-MRI) experiments have long reported solute transport in the brain that appears to be faster than diffusion alone, but this transport rate has not been quantified to a physically relevant value that can be compared to known diffusive rates of tracers. Methods In this work, DCE-MRI experimental data is analyzed using subject-specific finite-element models to quantify transport in different anatomical regions across the whole mouse brain. The set of regional effective diffusivities ($$D_{eff}$$ D eff ), a transport parameter combining all mechanisms of transport, that best represent the experimental data are determined and compared to apparent diffusivity ($$D_{app}$$ D app ), the known rate of diffusion through brain tissue, to draw conclusions about dominant transport mechanisms in each region. Results In the perivascular regions of major arteries, $$D_{eff}$$ D eff for gadoteridol (550 Da) was over 10,000 times greater than $$D_{app}$$ D app . In the brain tissue, constituting interstitial space and the perivascular space of smaller blood vessels, $$D_{eff}$$ D eff was 10–25 times greater than $$D_{app}$$ D app . Conclusions The analysis concludes that convection is present throughout the brain. Convection is dominant in the perivascular space of major surface and branching arteries (Pe > 1000) and significant to large molecules (> 1 kDa) in the combined interstitial space and perivascular space of smaller vessels (not resolved by DCE-MRI). Importantly, this work supports perivascular convection along penetrating blood vessels.
- Published
- 2021
- Full Text
- View/download PDF
14. Dynamic infrared imaging of cerebrospinal fluid tracer influx into the brain
- Author
-
Samantha A. Keil, Molly Braun, Ryan O’Boyle, Mathew Sevao, Taylor Pedersen, Sanjana Agarwal, Deidre Jansson, and Jeffrey J. Iliff
- Subjects
Radiological and Ultrasound Technology ,Neuroscience (miscellaneous) ,Radiology, Nuclear Medicine and imaging - Published
- 2021
15. Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid β plaque formation in mice
- Author
-
Matthew Simon, Marie Xun Wang, Ozama Ismail, Molly Braun, Abigail G. Schindler, Jesica Reemmer, Zhongya Wang, Mariya A. Haveliwala, Ryan P. O’Boyle, Warren Y. Han, Natalie Roese, Marjorie Grafe, Randall Woltjer, Detlev Boison, and Jeffrey J. Iliff
- Subjects
Aquaporin 4 ,Mice ,Amyloid beta-Peptides ,Neurology ,Alzheimer Disease ,Cognitive Neuroscience ,Animals ,Humans ,Plaque, Amyloid ,sense organs ,Neurology (clinical) ,Glymphatic System - Abstract
Background Slowed clearance of amyloid β (Aβ) is believed to underlie the development of Aβ plaques that characterize Alzheimer’s disease (AD). Aβ is cleared in part by the glymphatic system, a brain-wide network of perivascular pathways that supports the exchange of cerebrospinal and brain interstitial fluid. Glymphatic clearance, or perivascular CSF-interstitial fluid exchange, is dependent on the astroglial water channel aquaporin-4 (AQP4) as deletion of Aqp4 in mice slows perivascular exchange, impairs Aβ clearance, and promotes Aβ plaque formation. Methods To define the role of AQP4 in human AD, we evaluated AQP4 expression and localization in a human post mortem case series. We then used the α-syntrophin (Snta1) knockout mouse model which lacks perivascular AQP4 localization to evaluate the effect that loss of perivascular AQP4 localization has on glymphatic CSF tracer distribution. Lastly, we crossed this line into a mouse model of amyloidosis (Tg2576 mice) to evaluate the effect of AQP4 localization on amyloid β levels. Results In the post mortem case series, we observed that the perivascular localization of AQP4 is reduced in frontal cortical gray matter of subjects with AD compared to cognitively intact subjects. This decline in perivascular AQP4 localization was associated with increasing Aβ and neurofibrillary pathological burden, and with cognitive decline prior to dementia onset. In rodent studies, Snta1 gene deletion slowed CSF tracer influx and interstitial tracer efflux from the mouse brain and increased amyloid β levels. Conclusions These findings suggest that the loss of perivascular AQP4 localization may contribute to the development of AD pathology in human populations.
- Published
- 2021
16. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis
- Author
-
Susanna Törnroth-Horsefield, Mootaz M. Salman, Andrea M. Halsey, Alex C. Conner, Jerome Badaut, Marie Xun Wang, Roslyn M. Bill, Jeffrey J. Iliff, Philip Kitchen, Centre de résonance magnétique des systèmes biologiques (CRMSB), and Centre National de la Recherche Scientifique (CNRS)-Université de Bordeaux (UB)
- Subjects
Water flow ,[SDV]Life Sciences [q-bio] ,Aquaporin ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Homeostasis ,Humans ,030304 developmental biology ,Glia limitans ,Aquaporin 4 ,0303 health sciences ,Neurodegeneration ,Water ,Subcellular localization ,medicine.disease ,medicine.anatomical_structure ,Blood-Brain Barrier ,Astrocytes ,Glymphatic system ,Neurology (clinical) ,sense organs ,Neuroscience ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Aquaporin channels facilitate bidirectional water flow in all cells and tissues. AQP4 is highly expressed in astrocytes. In the CNS, it is enriched in astrocyte endfeet, at synapses, and at the glia limitans, where it mediates water exchange across the blood–spinal cord and blood–brain barriers (BSCB/BBB), and controls cell volume, extracellular space volume, and astrocyte migration. Perivascular enrichment of AQP4 at the BSCB/BBB suggests a role in glymphatic function. Recently, we have demonstrated that AQP4 localization is also dynamically regulated at the subcellular level, affecting membrane water permeability. Ageing, cerebrovascular disease, traumatic CNS injury, and sleep disruption are established and emerging risk factors in developing neurodegeneration, and in animal models of each, impairment of glymphatic function is associated with changes in perivascular AQP4 localization. CNS oedema is caused by passive water influx through AQP4 in response to osmotic imbalances. We have demonstrated that reducing dynamic relocalization of AQP4 to the BSCB/BBB reduces CNS oedema and accelerates functional recovery in rodent models. Given the difficulties in developing pore-blocking AQP4 inhibitors, targeting AQP4 subcellular localization opens up new treatment avenues for CNS oedema, neurovascular and neurodegenerative diseases, and provides a framework to address fundamental questions about water homeostasis in health and disease.
- Published
- 2021
- Full Text
- View/download PDF
17. MRI-Visible Perivascular Space (PVS) Changes with Long-Duration Spaceflight
- Author
-
Nichole E. Beltran, Rachael D. Seidler, S. Richmond, Jeffrey J. Iliff, Kathleen E. Hupfeld, Y. E. De Dios, Jacob J. Bloomberg, Heather R. McGregor, Juan Piantino, Madison Luther, Igor S. Kofman, Roy Riascos, Scott J. Wood, Lisa C. Silbert, Daniel Schwartz, and Ajitkumar P. Mulavara
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Automated segmentation ,Magnetic resonance imaging ,Spaceflight ,law.invention ,White matter ,medicine.anatomical_structure ,law ,Internal medicine ,cardiovascular system ,medicine ,Cardiology ,Ventricular volume ,Perivascular space ,Mri scan ,business ,Short duration - Abstract
Humans are exposed to extreme environmental stressors during spaceflight and return with alterations in brain structure and shifts in intracranial fluids. To date, no studies have evaluated the effects of spaceflight on perivascular spaces (PVSs) within the brain, which are believed to facilitate fluid drainage and brain homeostasis. Here, we examined how the number and morphology of magnetic resonance imaging (MRI)-visible PVSs are affected by spaceflight, including prior spaceflight experience. Fifteen astronauts underwent sixT1-weighted 3T MRI scans, twice prior to launch and four times following their return to Earth after ∼6-month missions to the International Space Station. White matter MRI-visible PVS number and morphology were calculated using an established automated segmentation algorithm. We found that novice astronauts showed an increase in total PVS volume from pre- to post-flight, whereas experienced crewmembers did not (adjusted for age, sex, and time between landing and first MRI scan). Moreover, experienced astronauts exhibited a significant correlation between more previous flight days and greater PVS median length at baseline, suggesting that experienced astronauts exhibit holdover effects from prior spaceflight(s). There was also a significant positive correlation between pre- to post-flight increases in PVS median length and increases in right lateral ventricular volume. The presence of spaceflight associated neuro-ocular syndrome (SANS) was not associated with PVS number or morphology. Together, these findings demonstrate that spaceflight is associated with PVS morphological changes, and specifically that spaceflight experience is an important factor in determining PVS characteristics.
- Published
- 2021
- Full Text
- View/download PDF
18. Longitudinal MRI-visible perivascular space (PVS) changes with long-duration spaceflight
- Author
-
Kathleen E, Hupfeld, Sutton B, Richmond, Heather R, McGregor, Daniel L, Schwartz, Madison N, Luther, Nichole E, Beltran, Igor S, Kofman, Yiri E, De Dios, Roy F, Riascos, Scott J, Wood, Jacob J, Bloomberg, Ajitkumar P, Mulavara, Lisa C, Silbert, Jeffrey J, Iliff, Rachael D, Seidler, and Juan, Piantino
- Subjects
Astronauts ,Humans ,Reproducibility of Results ,Space Flight ,Glymphatic System ,Magnetic Resonance Imaging - Abstract
Humans are exposed to extreme environmental stressors during spaceflight and return with alterations in brain structure and shifts in intracranial fluids. To date, no studies have evaluated the effects of spaceflight on perivascular spaces (PVSs) within the brain, which are believed to facilitate fluid drainage and brain homeostasis. Here, we examined how the number and morphology of magnetic resonance imaging (MRI)-visible PVSs are affected by spaceflight, including prior spaceflight experience. Fifteen astronauts underwent six T
- Published
- 2021
19. Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis
- Author
-
Mootaz M, Salman, Philip, Kitchen, Jeffrey J, Iliff, and Roslyn M, Bill
- Subjects
Aquaporin 4 ,Brain ,Homeostasis ,Humans - Published
- 2021
20. Link between Mild Traumatic Brain Injury, Poor Sleep, and Magnetic Resonance Imaging: Visible Perivascular Spaces in Veterans
- Author
-
Craig D. Newgard, Madison Luther, Juan Piantino, Natalia M. Kleinhans, Elaine R. Peskind, Jeffrey J. Iliff, Lisa C. Silbert, Daniel Schwartz, Murray A. Raskind, and Kathleen F. Pagulayan
- Subjects
Adult ,Male ,Sleep Wake Disorders ,030506 rehabilitation ,medicine.medical_specialty ,Traumatic brain injury ,Head trauma ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Perivascular space ,Brain trauma ,Iraq War, 2003-2011 ,Brain Concussion ,Veterans ,medicine.diagnostic_test ,Afghan Campaign 2001 ,business.industry ,Magnetic resonance imaging ,Original Articles ,medicine.disease ,Magnetic Resonance Imaging ,United States ,Poor sleep ,medicine.anatomical_structure ,Cardiology ,Glymphatic system ,Female ,Neurology (clinical) ,0305 other medical science ,business ,Glymphatic System ,030217 neurology & neurosurgery - Abstract
Impaired clearance of perivascular waste in the brain may play a critical role in morbidity after mild traumatic brain injury (mTBI). We aimed to determine the effect of mTBI on the burden of magnetic resonance imaging (MRI)-visible perivascular spaces (PVSs) in a cohort of U.S. military veterans and whether sleep modulates this effect. We also investigated the correlation between PVS burden and severity of persistent post-concussive symptoms. Fifty-six Iraq/Afghanistan veterans received 3 Tesla MRI as part of a prospective cohort study on military blast mTBI. White matter PVS burden (i.e., number and volume) was calculated using an established automated segmentation algorithm. Multi-variate regression was used to establish the association between mTBIs sustained in the military and PVS burden. Covariates included age, blood pressure, number of impact mTBIs outside the military, and blast exposures. Correlation coefficients were calculated between PVS burden and severity of persistent post-concussive symptoms. There was a significant positive relationship between the number of mTBIs sustained in the military and both PVS number and volume (p = 0.04). A significant interaction was found between mTBI and poor sleep on PVS volume (p = 0.04). A correlation was found between PVS number and volume, as well as severity of postconcussive symptoms (p = 0.03). Further analysis revealed a moderate correlation between PVS number and volume, as well as balance problems (p
- Published
- 2021
21. Vasomotor influences on glymphatic-lymphatic coupling and solute trafficking in the central nervous system
- Author
-
Jeffrey J. Iliff and James R. Goodman
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Meningeal lymphatic vessels ,Central nervous system ,Contrast Media ,Hypercapnia ,Mice ,03 medical and health sciences ,Meninges ,0302 clinical medicine ,Cerebrospinal fluid ,medicine ,Animals ,Anesthesia ,Cerebrospinal Fluid ,030304 developmental biology ,0303 health sciences ,Vasomotor ,Chemistry ,Respiration ,Brain ,Extracellular Fluid ,Original Articles ,Respiration, Artificial ,Mice, Inbred C57BL ,Coupling (electronics) ,Lymphatic system ,medicine.anatomical_structure ,Neurology ,Cerebrovascular Circulation ,Female ,Glymphatic system ,Lymph Nodes ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,Glymphatic System ,030217 neurology & neurosurgery - Abstract
Despite the recent description of meningeal lymphatic vessels draining solutes from the brain interstitium and cerebrospinal fluid (CSF), the physiological factors governing cranial lymphatic efflux remain largely unexplored. In agreement with recent findings, cervical lymphatic drainage of 70 kD and 2000 kD fluorescent tracers injected into the adult mouse cortex was significantly impaired in the anesthetized compared to waking animals (tracer distribution across 2.1 ± 4.5% and 23.7 ± 15.8% of deep cervical lymph nodes, respectively); however, free-breathing anesthetized mice were markedly hypercapnic and acidemic (paCO2 = 64 ± 8 mmHg; pH = 7.22 ± 0.05). Mechanical ventilation normalized arterial blood gases in anesthetized animals, and rescued lymphatic efflux of interstitial solutes in anesthetized mice. Experimental hypercapnia blocked cervical lymphatic efflux of intraparenchymal tracers. When tracers were injected into the subarachnoid CSF compartment, glymphatic influx into brain tissue was virtually abolished by hypercapnia, while lymphatic drainage was not appreciably altered. These findings demonstrate that cervical lymphatic drainage of interstitial solutes is, in part, regulated by upstream changes in glymphatic CSF-interstitial fluid exchange. Further, they suggest that maintaining physiological blood gas values in studies of glymphatic exchange and meningeal lymphatic drainage may be critical to defining the physiological regulation of these processes.
- Published
- 2019
- Full Text
- View/download PDF
22. Pericyte constriction underlies capillary derecruitment during hyperemia in the setting of arterial stenosis
- Author
-
Wei Wei, Nabil J. Alkayed, Carmen Methner, Sanjiv Kaul, Ruikang K. Wang, N. David Yanez, Anusha Mishra, Jeffrey J. Iliff, Yuandong Li, Kirsti Golgotiu, and Berislav V. Zlokovic
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Ischemia ,Hyperemia ,Mice, Transgenic ,Constriction, Pathologic ,Constriction ,Receptor, Platelet-Derived Growth Factor beta ,Mice ,Peripheral Arterial Disease ,Physiology (medical) ,Internal medicine ,Animals ,Medicine ,Ligation ,business.industry ,Arterial stenosis ,Cardiac ischemia ,Angiography ,medicine.disease ,Capillaries ,Femoral Artery ,Vasodilation ,Disease Models, Animal ,Luminescent Proteins ,Stenosis ,Microscopy, Fluorescence, Multiphoton ,medicine.anatomical_structure ,Regional Blood Flow ,Vasoconstriction ,Gracilis Muscle ,Mutation ,cardiovascular system ,Cardiology ,Female ,Pericyte ,Pericytes ,Cardiology and Cardiovascular Medicine ,business ,Perfusion ,Research Article ,circulatory and respiratory physiology - Abstract
Capillary derecruitment distal to a coronary stenosis is implicated as the mechanism of reversible perfusion defect and potential myocardial ischemia during coronary hyperemia; however, the underlying mechanisms are not defined. We tested whether pericyte constriction underlies capillary derecruitment during hyperemia under conditions of stenosis. In vivo two-photon microscopy (2PM) and optical microangiography (OMAG) were used to measure hyperemia-induced changes in capillary diameter and perfusion in wild-type and pericyte-depleted mice with femoral artery stenosis. OMAG demonstrated that hyperemic challenge under stenosis produced capillary derecruitment associated with decreased RBC flux. 2PM demonstrated that hyperemia under control conditions induces 26 ± 5% of capillaries to dilate and 19 ± 3% to constrict. After stenosis, the proportion of capillaries dilating to hyperemia decreased to 14 ± 4% ( P = 0.05), whereas proportion of constricting capillaries increased to 32 ± 4% ( P = 0.05). Hyperemia-induced changes in capillary diameter occurred preferentially in capillary segments invested with pericytes. In a transgenic mouse model featuring partial pericyte depletion, only 14 ± 3% of capillaries constricted to hyperemic challenge after stenosis, a significant reduction from 33 ± 4% in wild-type littermate controls ( P = 0.04). These results provide for the first time direct visualization of hyperemia-induced capillary derecruitment distal to arterial stenosis and demonstrate that pericyte constriction underlies this phenomenon in vivo. These results could have important therapeutic implications in the treatment of exercise-induced ischemia. NEW & NOTEWORTHY In the setting of coronary arterial stenosis, hyperemia produces a reversible perfusion defect resulting from capillary derecruitment that is believed to underlie cardiac ischemia under hyperemic conditions. We use optical microangiography and in vivo two-photon microscopy to visualize capillary derecruitment distal to a femoral arterial stenosis with cellular resolution. We demonstrate that capillary constriction in response to hyperemia in the setting of stenosis is dependent on pericytes, contractile mural cells investing the microcirculation.
- Published
- 2019
- Full Text
- View/download PDF
23. Sleep as a Therapeutic Target in the Aging Brain
- Author
-
James R. Goodman, Thierno Madjou Bah, and Jeffrey J. Iliff
- Subjects
0301 basic medicine ,Aging ,medicine.medical_specialty ,Neurology ,Review ,Disease ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Dementia ,Aging brain ,Pharmacology (medical) ,Cognitive decline ,Pharmacology ,business.industry ,Brain ,Neurodegenerative Diseases ,medicine.disease ,Sleep time ,Sleep in non-human animals ,Pathophysiology ,030104 developmental biology ,Neurology (clinical) ,Sleep ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Sleep is a behavioral phenomenon conserved among mammals and some invertebrates, yet the biological functions of sleep are still being elucidated. In humans, sleep time becomes shorter, more fragmented, and of poorer quality with advancing age. Epidemiologically, the development of age-related neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease is associated with pronounced sleep disruption, whereas emerging mechanistic studies suggest that sleep disruption may be causally linked to neurodegenerative pathology, suggesting that sleep may represent a key therapeutic target in the prevention of these conditions. In this review, we discuss the physiology of sleep, the pathophysiology of neurodegenerative disease, and the current literature supporting the relationship between sleep, aging, and neurodegenerative disease. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s13311-019-00769-6) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
24. Pulsation changes link to impaired glymphatic function in a mouse model of vascular cognitive impairment
- Author
-
Beverley J, Jeffrey J. Iliff, Roxana O. Carare, Jessica Duncombe, Raj N. Kalaria, Wiegand Uk, Akihiro Kitamura, Mengxi Li, Bettina Platt, Karen Horsburgh, and Juraj Koudelka
- Subjects
medicine.medical_specialty ,Amyloid ,business.industry ,medicine.disease ,Pathophysiology ,Astrogliosis ,Cortex (botany) ,Cerebrospinal fluid ,Internal medicine ,medicine ,Cardiology ,Glymphatic system ,Cerebral perfusion pressure ,Beta (finance) ,business - Abstract
Large vessel disease and carotid stenosis are key mechanisms contributing to vascular cognitive impairment (VCI) and dementia. Our previous work, and that of others, using rodent models, demonstrated that bilateral common carotid stenosis (BCAS) leads to cognitive impairment via gradual deterioration of the glial-vascular unit and accumulation of amyloid-β (Aβ) protein. Since brain-wide drainage pathways (glymphatic) for waste clearance, including Aβ removal, have been implicated in the pathophysiology of VCI via glial mechanisms, we hypothesized that glymphatic function would be impaired in a BCAS model and exacerbated in the presence of Aβ. Male wild- type and Tg-SwDI (model of microvascular amyloid) mice were subjected to BCAS or sham surgery which led to a reduction in cerebral perfusion and impaired spatial learning and memory. After 3 months survival, glymphatic function was evaluated by cerebrospinal fluid (CSF) fluorescent tracer influx. We demonstrated that BCAS caused a marked regional reduction of CSF tracer influx in the dorsolateral cortex and CA1-DG molecular layer. In parallel to these changes increased reactive astrogliosis was observed post-BCAS. To further investigate the mechanisms that may lead to these changes, we measured the pulsation of cortical vessels. BCAS impaired vascular pulsation in pial arteries in WT and Tg-SwDI mice. Since our findings show that BCAS may influence VCI by impaired glymphatic drainage and reduced vascular pulsation we propose that these additional targets need to be considered when treating VCI.
- Published
- 2021
- Full Text
- View/download PDF
25. Woes with flows under pressure
- Author
-
Deidre Jansson and Jeffrey J. Iliff
- Subjects
Nerve degeneration ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,AcademicSubjects/SCI01870 ,glymphatic system ,Pseudotumor cerebri ,business.industry ,General Engineering ,Ischemia ,Magnetic resonance imaging ,Brain tissue ,medicine.disease ,cerebrospinal fluid ,medicine ,Original Article ,AcademicSubjects/MED00310 ,Glymphatic system ,pulsatile intracranial pressure ,business ,idiopathic intracranial hypertension ,MRI ,Clearance - Abstract
Idiopathic intracranial hypertension is a brain disease incorporating cerebrospinal fluid disturbance, increased intracranial pressure and visual failure, but with unknown cause. This study examined a hypothesis that glymphatic function is impaired in idiopathic intracranial hypertension patients. The MRI contrast agent gadobutrol was utilized as a cerebrospinal fluid tracer following intrathecal administration. Consecutive standardized T1 MRI acquisitions over 48 h were done to assess tracer distribution within brain of 15 idiopathic intracranial hypertension patients and 15 reference individuals who were comparable in age and gender distribution. Using FreeSurfer software, we semi-quantified tracer level in multiple brain regions as T1 MRI signal change. The tracer enriched the entire brain of idiopathic intracranial hypertension and reference subjects. In idiopathic intracranial hypertension, tracer enrichment was increased and clearance of tracer delayed from a wide range of brain regions, including both grey and white matter. Differences were most evident in frontal and temporal regions. The pulsatile intracranial pressure was measured overnight and tracer propagation in brain compared between individuals with pathological and normal pulsatile intracranial pressure. In individuals with pathological pulsatile intracranial pressure, tracer enrichment was stronger and clearance from brain delayed, particularly in regions nearby large artery trunks at the brain surface. The present in vivo observations provide evidence for impaired glymphatic function in several brain regions of idiopathic intracranial hypertension patients. Glymphatic failure may imply altered clearance of metabolic byproducts, which may precede neurodegeneration. Further studies are needed to characterize glymphatic failure in idiopathic intracranial hypertension., Intrathecal contrast-enhanced MRI provided evidence for impaired glymphatic function in many brain regions, including both grey and white matter, of patients with idiopathic intracranial hypertension, a brain disease characterized by CSF disturbance and increased intracranial pressure of unknown cause., Graphical Abstract Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
26. Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis
- Author
-
Mootaz M. Salman, Jeffrey J. Iliff, Philip Kitchen, and Roslyn M. Bill
- Subjects
medicine.anatomical_structure ,Aquaporin 4 ,Cellular neuroscience ,Fluid homeostasis ,Chemistry ,General Neuroscience ,medicine ,Glymphatic system ,Blood–brain barrier ,Neuroscience - Published
- 2021
- Full Text
- View/download PDF
27. Varying perivascular astroglial endfoot dimensions along the vascular tree maintain perivascular-interstitial flux through the cortical mantle
- Author
-
Kenji F. Tanaka, Jeffrey J. Iliff, Marie Xun Wang, Lori Ray, and Jeffrey J. Heys
- Subjects
0301 basic medicine ,Large vessel ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,medicine ,Animals ,Perivascular space ,Mantle (mollusc) ,Chemistry ,Brain ,Membrane Proteins ,Anatomy ,Immunofluorescent labeling ,Fluid exchange ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Physical Barrier ,Astrocytes ,Marked heterogeneity ,Glymphatic system ,030217 neurology & neurosurgery ,Astrocyte - Abstract
The glymphatic system is a recently defined brain-wide network of perivascular spaces along which cerebrospinal fluid (CSF) and interstitial solutes exchange. Astrocyte endfeet encircling the perivascular space form a physical barrier in between these two compartments, and fluid and solutes that are not taken up by astrocytes move out of the perivascular space through the junctions in between astrocyte endfeet. However, little is known about the anatomical structure and the physiological roles of the astrocyte endfeet in regulating the local perivascular exchange. Here, visualizing astrocyte endfoot-endfoot junctions with immunofluorescent labeling against the protein megalencephalic leukoencephalopathy with subcortical cysts-1 (MLC1), we characterized endfoot dimensions along the mouse cerebrovascular tree. We observed marked heterogeneity in endfoot dimensions along vessels of different sizes, and of different types. Specifically, endfoot size was positively correlated with the vessel diameters, with large vessel segments surrounded by large endfeet and small vessel segments surrounded by small endfeet. This association was most pronounced along arterial, rather than venous segments. Computational modeling simulating vascular trees with uniform or varying endfeet dimensions demonstrates that varying endfoot dimensions maintain near constant perivascular-interstitial flux despite correspondingly declining perivascular pressures along the cerebrovascular tree through the cortical depth. These results describe a novel anatomical feature of perivascular astroglial endfeet and suggest that endfoot heterogeneity may be an evolutionary adaptation to maintain perivascular CSF-interstitial fluid exchange through deep brain structures.
- Published
- 2020
28. The impact of neurovascular, blood-brain barrier, and glymphatic dysfunction in neurodegenerative and metabolic diseases
- Author
-
Molly, Braun and Jeffrey J, Iliff
- Subjects
Aging ,Diabetes Mellitus, Type 2 ,Blood-Brain Barrier ,Cerebrovascular Circulation ,Choroid Plexus ,Animals ,Humans ,Neurodegenerative Diseases ,Glymphatic System ,Cerebrospinal Fluid - Abstract
The cerebral vasculature serves as the crossroads of the CNS, supporting exchange of nutrients, metabolic wastes, solutes and cells between the compartments of the brain, including the blood, brain interstitium, and cerebrospinal fluid (CSF). The blood-brain barrier (BBB) regulates the entry and efflux of molecules into brain tissue. The cells of the neurovascular unit regulate cerebral blood flow, matching local metabolic demand to blood supply. The blood-CSF barrier at the choroid plexus secretes CSF, which supports the brain and provides a sink for interstitial solutes not cleared across the BBB. Recent studies have characterized the glymphatic system, a brain-wide network of perivascular spaces that supports CSF and interstitial fluid exchange and the clearance of interstitial solutes to the CSF. The critical role that these structures play in maintaining brain homeostasis is illustrated by the established and emerging roles that their dysfunctions play in the development of neurodegenerative diseases, such as Alzheimer's disease (AD). Loss of BBB and blood-CSF barrier function is reported both in rodent models of AD, and in human AD subjects. Cerebrovascular dysfunction and ischemic injury are well established contributors to both vascular dementia and to a large proportion of cases of sporadic AD. In animal models, the slowed glymphatic clearance of interstitial proteins, such as amyloid β or tau, are proposed to contribute to the development of neurodegenerative diseases, including AD. In total, these findings suggest that cellular and molecular changes occurring within and around the cerebral vasculature are among the key drivers of neurodegenerative disease pathogenesis.
- Published
- 2020
29. The impact of neurovascular, blood-brain barrier, and glymphatic dysfunction in neurodegenerative and metabolic diseases
- Author
-
Jeffrey J. Iliff and Molly Braun
- Subjects
biology ,Amyloid beta ,business.industry ,Blood–brain barrier ,03 medical and health sciences ,Cerebral circulation ,0302 clinical medicine ,Cerebrospinal fluid ,medicine.anatomical_structure ,Cerebral blood flow ,medicine ,biology.protein ,Choroid plexus ,Glymphatic system ,Perivascular space ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The cerebral vasculature serves as the crossroads of the CNS, supporting exchange of nutrients, metabolic wastes, solutes and cells between the compartments of the brain, including the blood, brain interstitium, and cerebrospinal fluid (CSF). The blood-brain barrier (BBB) regulates the entry and efflux of molecules into brain tissue. The cells of the neurovascular unit regulate cerebral blood flow, matching local metabolic demand to blood supply. The blood-CSF barrier at the choroid plexus secretes CSF, which supports the brain and provides a sink for interstitial solutes not cleared across the BBB. Recent studies have characterized the glymphatic system, a brain-wide network of perivascular spaces that supports CSF and interstitial fluid exchange and the clearance of interstitial solutes to the CSF. The critical role that these structures play in maintaining brain homeostasis is illustrated by the established and emerging roles that their dysfunctions play in the development of neurodegenerative diseases, such as Alzheimer's disease (AD). Loss of BBB and blood-CSF barrier function is reported both in rodent models of AD, and in human AD subjects. Cerebrovascular dysfunction and ischemic injury are well established contributors to both vascular dementia and to a large proportion of cases of sporadic AD. In animal models, the slowed glymphatic clearance of interstitial proteins, such as amyloid β or tau, are proposed to contribute to the development of neurodegenerative diseases, including AD. In total, these findings suggest that cellular and molecular changes occurring within and around the cerebral vasculature are among the key drivers of neurodegenerative disease pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
30. Transcriptional signatures in histologic structures within glioblastoma tumors may predict personalized drug sensitivity and survival
- Author
-
Jeffrey J. Iliff, Joe W. Gray, Daniel Schwartz, James E. Korkola, Tyler Risom, Cheryl J. Claunch, Elmar Bucher, Laura M. Heiser, Ramon F. Barajas, Leslie L. Muldoon, Prakash Ambady, Edward A. Neuwelt, and Cymon Kersch
- Subjects
0301 basic medicine ,Oncology ,Drug ,medicine.medical_specialty ,media_common.quotation_subject ,Biology ,gene signature ,Transcriptome ,transcriptomics ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Cancer genome ,medicine ,AcademicSubjects/MED00300 ,Survival analysis ,media_common ,glioblastoma ,Gene signature ,medicine.disease ,Subtyping ,Gene expression profiling ,030104 developmental biology ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,AcademicSubjects/MED00310 ,heterogeneity ,Glioblastoma - Abstract
Background Glioblastoma is a rapidly fatal brain cancer that exhibits extensive intra- and intertumoral heterogeneity. Improving survival will require the development of personalized treatment strategies that can stratify tumors into subtypes that differ in therapeutic vulnerability and outcomes. Glioblastoma stratification has been hampered by intratumoral heterogeneity, limiting our ability to compare tumors in a consistent manner. Here, we develop methods that mitigate the impact of intratumoral heterogeneity on transcriptomic-based patient stratification. Methods We accessed open-source transcriptional profiles of histological structures from 34 human glioblastomas from the Ivy Glioblastoma Atlas Project. Principal component and correlation network analyses were performed to assess sample inter-relationships. Gene set enrichment analysis was used to identify enriched biological processes and classify glioblastoma subtype. For survival models, Cox proportional hazards regression was utilized. Transcriptional profiles from 156 human glioblastomas were accessed from The Cancer Genome Atlas to externally validate the survival model. Results We showed that intratumoral histologic architecture influences tumor classification when assessing established subtyping and prognostic gene signatures, and that indiscriminate sampling can produce misleading results. We identified the cellular tumor as a glioblastoma structure that can be targeted for transcriptional analysis to more accurately stratify patients by subtype and prognosis. Based on expression from cellular tumor, we created an improved risk stratification gene signature. Conclusions Our results highlight that biomarker performance for diagnostics, prognostics, and prediction of therapeutic response can be improved by analyzing transcriptional profiles in pure cellular tumor, which is a critical step toward developing personalized treatment for glioblastoma.
- Published
- 2020
- Full Text
- View/download PDF
31. Targeted Assessment of Enlargement of the Perivascular Space in Alzheimer’s Disease and Vascular Dementia Subtypes Implicates Astroglial Involvement Specific to Alzheimer’s Disease
- Author
-
Erin L. Boespflug, Jeffrey J. Iliff, Matthew J. Simon, Emmalyn Leonard, Marjorie R. Grafe, Randall L. Woltjer, Lisa C. Silbert, and Jeffrey Kaye
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Disease ,Article ,03 medical and health sciences ,Cerebral circulation ,0302 clinical medicine ,Alzheimer Disease ,Parenchyma ,Humans ,Medicine ,Perivascular space ,Vascular dementia ,Aged ,Aged, 80 and over ,Aquaporin 4 ,business.industry ,Dementia, Vascular ,General Neuroscience ,Brain ,General Medicine ,Middle Aged ,medicine.disease ,Psychiatry and Mental health ,Clinical Psychology ,030104 developmental biology ,medicine.anatomical_structure ,Water channel ,Astrocytes ,Female ,Glymphatic system ,Geriatrics and Gerontology ,business ,Glymphatic System ,030217 neurology & neurosurgery - Abstract
Waste clearance from the brain parenchyma occurs along perivascular pathways. Enlargement of the perivascular space (ePVS) is associated with pathologic features of Alzheimer's disease (AD), although the mechanisms and implications of this dilation are unclear. Fluid exchange along the cerebral vasculature is dependent on the perivascular astrocytic water channel aquaporin-4 (AQP4) and loss of perivascular AQP4 localization is found in AD. We directly measured ePVS in postmortem samples of pathologically characterized tissue from participants who were cognitively intact or had AD or mixed dementia (vascular lesions with AD). We found that both AD and mixed dementia groups had significantly increased ePVS compared to cognitively intact subjects. In addition, we found increased global AQP4 expression of the AD group over both control and mixed dementia groups and a qualitative reduction in perivascular localization of AQP4 in the AD group. Among these cases, increasing ePVS burden was associated with the presence of tau and amyloid-β pathology. These findings are consistent with the existing evidence of ePVS in AD and provide novel information regarding differences in AD and vascular dementia and the potential role of astroglial pathology in ePVS.
- Published
- 2018
- Full Text
- View/download PDF
32. 252 Non-Invasive Quantification of Human Brain Lactate Concentrations Across Sleep-Wake Cycles
- Author
-
Miranda M. Lim, Manoj K. Sammi, Charles F. Murchison, Selda Yildiz, Katherine Powers, Jeffrey J. Iliff, and William D. Rooney
- Subjects
medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Physiology (medical) ,Internal medicine ,Sleep wake ,Non invasive ,Cardiology ,medicine ,Neurology (clinical) ,Human brain ,business - Abstract
Introduction Cellular mechanisms underlying changes in small animal brain lactate concentrations have been investigated for more than 70 years and report sharp reductions in lactate (12-35%) during sleep or anesthesia relative to wakefulness. The goal of this study was to investigate alterations in human cerebral lactate concentrations across sleep-wake cycles. Toward this goal, we developed a novel non-invasive methodology, quantified changes in human cerebral lactate during sleep stages, and investigated potential mechanisms associated with changes in lactate. Methods Nine subjects (four females, five males; 21-27 y-o, mean age 24.2 ±2) were sleep deprived overnight, and underwent (5:45~11:00 am) experiments combining simultaneous MR-spectroscopy (MRS) and polysomnography (PSG) in a 3 T MR instrument using a 64-channel head/neck coil. A single voxel MRS (1H-MRS) acquired signals from a volume of interest (12~24 cm3) for every 7.5-s for 88~180-min. Lactate signal intensity was determined from each 7.5-s spectrum, normalized to corresponding water signal, and averaged over 30-s for each PSG epochs. Artifact corrected PSG data were scored for each 30-s epoch using the standard criteria and classified into one of four stages: W, N1, N2 and N3. Group mean lactate levels were quantified using LCModel. Three subjects returned for lactate diffusivity measurements using diffusion-sensitized PRESS MRS sequence. Results Compared to W, group mean lactate levels within each sleep stage showed a reduction of [4.9 ± 4.9] % in N1, [10.4 ± 5.2] % in N2, and [24.0 ± 5.8] % in N3. We observed a significant decrease in lactate apparent diffusion coefficient (ADC) accompanied by reduced brain lactate in sleep compared to wake (P Conclusion This is the first in-vivo report of alterations in human brain lactate concentrations across sleep-wake cycles. Observed decline in lactate levels during sleep compared to wakefulness is consistent with, and extends results from invasive small animal brain studies first reported more than 70 years ago, and support the notion of altered lactate metabolism and/or increased glymphatic activity in sleeping human brain. Support (if any) The Paul. G. Allen Family Foundation funded the study.
- Published
- 2021
- Full Text
- View/download PDF
33. Neurovascular Unit: Basic and Clinical Imaging with Emphasis on Advantages of Ferumoxytol
- Author
-
Bronwyn E. Hamilton, Ashok Panigrahy, Heike E. Daldrup-Link, Edward A. Neuwelt, Csanad Varallyay, Danica Stanimirovic, Seymur Gahramanov, Saeid Taheri, Haris I. Sair, Christopher D d'Esterre, Daniel S. Reich, Jeffrey J. Iliff, Joao Prola Netto, Mauricio Castillo, Kenneth A. Krohn, Lester R. Drewes, Yueh Z. Lee, Rajan Jain, and Berislav V. Zlokovic
- Subjects
Central nervous system ,Contrast Media ,Metal Nanoparticles ,Neuroimaging ,Vascular permeability ,Review ,Blood–brain barrier ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,neurovascular unit ,Tight junction ,business.industry ,imaging ,blood-brain barrier ,Neurovascular bundle ,Ferrosoferric Oxide ,Ferumoxytol ,Functional imaging ,medicine.anatomical_structure ,Surgery ,Basal lamina ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Physiological and pathological processes that increase or decrease the central nervous system's need for nutrients and oxygen via changes in local blood supply act primarily at the level of the neurovascular unit (NVU). The NVU consists of endothelial cells, associated blood–brain barrier tight junctions, basal lamina, pericytes, and parenchymal cells, including astrocytes, neurons, and interneurons. Knowledge of the NVU is essential for interpretation of central nervous system physiology and pathology as revealed by conventional and advanced imaging techniques. This article reviews current strategies for interrogating the NVU, focusing on vascular permeability, blood volume, and functional imaging, as assessed by ferumoxytol an iron oxide nanoparticle.
- Published
- 2017
- Full Text
- View/download PDF
34. The effects of noncoding aquaporin-4 single-nucleotide polymorphisms on cognition and functional progression of Alzheimer's disease
- Author
-
Charles Murchison, Jeffrey J. Iliff, Jeffrey Kaye, Joseph F. Quinn, Kevin G. Burfeind, Deniz Erten-Lyons, Matthew J. Simon, and Shawn K. Westaway
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cognitive decline ,Single-nucleotide polymorphism ,Disease ,Biology ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Genetics ,Aging brain ,Dementia ,Senile plaques ,Amyloid β ,Featured Article ,Alzheimer's disease ,medicine.disease ,Psychiatry and Mental health ,030104 developmental biology ,Aquaporin 4 ,Glymphatic system ,Neurology (clinical) ,sense organs ,Cohort study ,Aquaporin-4 ,030217 neurology & neurosurgery - Abstract
Introduction The glymphatic system is a brain-wide perivascular network that facilitates clearance of proteins, including amyloid β, from the brain interstitium through the perivascular exchange of cerebrospinal fluid and interstitial fluid. The astrocytic water channel aquaporin-4 (AQP4) is required for glymphatic system function, and impairment of glymphatic function in the aging brain is associated with altered AQP4 expression and localization. In human cortical tissue, alterations in AQP4 expression and localization are associated with Alzheimer's disease (AD) status and pathology. Although this suggests a potential role for AQP4 in the development or progression of AD, the relationship between of naturally occurring variants in the human AQP4 gene and cognitive function has not yet been evaluated. Methods Using data from several longitudinal aging cohorts, we investigated the association between five AQP4 single-nucleotide polymorphisms (SNPs) and the rate of cognitive decline in participants with a diagnosis of AD. Results None of the five SNPs were associated with different rates of AD diagnosis, age of dementia onset in trial subjects. No association between AQP4 SNPs with histological measures of AD pathology, including Braak stage or neuritic plaque density was observed. However, AQP4 SNPs were associated with altered rates of cognitive decline after AD diagnosis, with two SNPS (rs9951307 and rs3875089) associated with slower cognitive decline and two (rs3763040 and rs3763043) associated with more rapid cognitive decline after AD diagnosis. Discussion These results provide the first evidence that variations in the AQP4 gene, whose gene product AQP4 is vital for glymphatic pathway function, may modulate the progression of cognitive decline in AD.
- Published
- 2017
35. Rebuttal from Matthew Simon and Jeffrey Iliff
- Author
-
Matthew J. Simon and Jeffrey J. Iliff
- Subjects
Aquaporin 4 ,Diffusion ,Physiology ,Philosophy ,Rebuttal ,Brain ,Theology ,Article - Published
- 2019
36. The glymphatic system supports convective exchange of cerebrospinal fluid and brain interstitial fluid that is mediated by perivascular aquaporin-4
- Author
-
Jeffrey J. Iliff and Matthew J. Simon
- Subjects
Aquaporin 4 ,Physiology ,Chemistry ,Brain ,Extracellular Fluid ,Article ,Crosstalk (biology) ,Cerebrospinal fluid ,Interstitial fluid ,Biophysics ,Animals ,Humans ,Glymphatic system ,Glymphatic System ,Cerebrospinal Fluid - Published
- 2019
37. Linking Traumatic Brain Injury, Sleep Disruption and Post-Traumatic Headache: a Potential Role for Glymphatic Pathway Dysfunction
- Author
-
Jeffrey J. Iliff, Juan Piantino, Craig D. Newgard, and Miranda M. Lim
- Subjects
Sleep Wake Disorders ,Traumatic brain injury ,Disrupted sleep ,Dysfunctional family ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,030202 anesthesiology ,Concussion ,Brain Injuries, Traumatic ,medicine ,Humans ,business.industry ,General Medicine ,medicine.disease ,Sleep in non-human animals ,Anesthesiology and Pain Medicine ,Post-Traumatic Headache ,Glymphatic system ,Neurology (clinical) ,Headaches ,medicine.symptom ,business ,Glymphatic System ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Traumatic brain injury (TBI) is a major public health concern in the USA and worldwide. Sleep disruption and headaches are two of the most common problems reported by patients after TBI. In this manuscript, we review the current knowledge regarding the relation between post-traumatic sleep disruption and headaches. We also describe the role of the glymphatic system as a potential link between TBI, sleep, and headaches. Recent studies show a reciprocal relation between post-traumatic sleep disruption and headaches: patients with sleep disruption after TBI report more headaches, and post-traumatic headaches are a risk factor for developing disrupted sleep. Despite this clinical association, the exact mechanisms linking post-traumatic sleep disruption and headaches are not well understood. The glymphatic pathway, a newly described brain–wide network of perivascular spaces that supports the clearance of interstitial solutes and wastes from the brain, is active primarily during sleep, and becomes dysfunctional after TBI. We propose a model where changes in glymphatic function caused by TBI and post-traumatic sleep disruption may impair the clearance of neuropeptides involved in the pathogenesis of post-traumatic headaches, such as CGRP. The relation between TBI, post-traumatic sleep disruption, and post-traumatic headaches, although well documented in the literature, remains poorly understood. Dysfunction of the glymphatic system caused by TBI offers a novel and exiting explanation to this clinically observed phenomenon. The proposed model, although theoretical, could provide important mechanistic insights to the TBI-sleep-headache association.
- Published
- 2019
38. In response to 'Is solute movement within the extracellular spaces of brain gray matter brought about primarily by diffusion or flow?'
- Author
-
Lori Ray, Jeffrey J. Heys, and Jeffrey J. Iliff
- Subjects
0301 basic medicine ,Convection ,Work (thermodynamics) ,Flow (psychology) ,lcsh:RC346-429 ,Diffusion ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Range (statistics) ,Humans ,Diffusion (business) ,Gray Matter ,lcsh:Neurology. Diseases of the nervous system ,Physics ,Superficial velocity ,Brain ,Biological Transport ,General Medicine ,Mechanics ,Letter to the Editor Response ,Volumetric flow rate ,030104 developmental biology ,Neurology ,Volume (thermodynamics) ,Extracellular Space ,030217 neurology & neurosurgery - Abstract
In our work, “Analysis of Convective and Diffusive Transport in the Brain Interstitium”, published in this journal (2019, 16:6), we estimate the interstitial superficial velocity by comparison of transport model simulations to published experimental Real-Time Iontophoresis (RTI) data. In the Discussion section, we calculate a value for perfusion rate, or volumetric flow rate per unit mass of tissue, from these fundamental results of superficial velocity. Drs. Hladky and Barrand have proposed an alternative method for choosing the surface area per volume used to calculate perfusion rate from superficial velocity, using our model domain. Their method seems reasonable to us, as does ours. Upon reflection, a range of volumetric flow per unit mass values should have been reported in our paper, 1–40 μL/min-g. The value calculated using Drs. Hladky and Barrand surface area is a likely upper-bound on this range and the value in the paper is a low estimate at the bottom of the range. We are confident in the estimates of interstitial velocity reported in our article, using the assumptions of the model. Peclet (Pe) numbers, which compare convective and diffusive transport rates for different molecules, were calculated using the superficial velocity estimates; and we continue to believe these values are correct along with all other major results and conclusions presented in the paper.
- Published
- 2019
39. Structural and functional conservation of non-lumenized lymphatic endothelial cells in the mammalian leptomeninges
- Author
-
Shannon Shibata-Germanos, Alan Grieg, James R. Goodman, Jason Rihel, Sandrine C. Foti, Tammaryn Lashley, Thomas A. Hawkins, Raphael F. P. Castellan, Roy O. Weller, Jeffrey J. Iliff, Bridget C. Benson, Rosa Maria Correra, Melissa Barber, Christiana Ruhrberg, Ana Faro, and Chintan A. Trivedi
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Cell type ,CNS lymphatics ,Dura mater ,government.form_of_government ,Biology ,Pathology and Forensic Medicine ,Lymphatic System ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Cerebrospinal fluid ,Meninges ,medicine ,Animals ,Humans ,Zebrafish ,030304 developmental biology ,Aged ,Aged, 80 and over ,Meningeal lymphatics ,0303 health sciences ,Original Paper ,Amyloid beta-Peptides ,Leptomeninges ,Macrophages ,Brain ,Endothelial Cells ,biology.organism_classification ,Lymphatic Endothelium ,medicine.anatomical_structure ,Lymphatic system ,government ,Lymphatics ,Female ,Neurology (clinical) ,CNS macrophages ,030217 neurology & neurosurgery - Abstract
The vertebrate CNS is surrounded by the meninges, a protective barrier comprised of the outer dura mater and the inner leptomeninges, which includes the arachnoid and pial layers. While the dura mater contains lymphatic vessels, no conventional lymphatics have been found within the brain or leptomeninges. However, non-lumenized cells called Brain/Mural Lymphatic Endothelial Cells or Fluorescent Granule Perithelial cells (muLECs/BLECs/FGPs) that share a developmental program and gene expression with peripheral lymphatic vessels have been described in the meninges of zebrafish. Here we identify a structurally and functionally similar cell type in the mammalian leptomeninges that we name Leptomeningeal Lymphatic Endothelial Cells (LLEC). As in zebrafish, LLECs express multiple lymphatic markers, containing very large, spherical inclusions, and develop independently from the meningeal macrophage lineage. Mouse LLECs also internalize macromolecules from the cerebrospinal fluid, including Amyloid-β, the toxic driver of Alzheimer’s disease progression. Finally, we identify morphologically similar cells co-expressing LLEC markers in human post-mortem leptomeninges. Given that LLECs share molecular, morphological, and functional characteristics with both lymphatics and macrophages, we propose they represent a novel, evolutionary conserved cell type with potential roles in homeostasis and immune organization of the meninges. Electronic supplementary material The online version of this article (10.1007/s00401-019-02091-z) contains supplementary material, which is available to authorized users.
- Published
- 2019
40. Role of endothelium-pericyte signaling in capillary blood flow response to neuronal activity
- Author
-
Douglas M. Zeppenfeld, Nabil J. Alkayed, Mariya Haveliwala, Kirsti Golgotiu, Daniel Hong, Jeffrey J. Iliff, Wenri Zhang, Marie Xun Wang, Ruikang K. Wang, Yuandong Li, and Catherine M. Davis
- Subjects
Male ,Endothelium ,Hyperemia ,Stimulation ,Epoxyeicosatrienoic acid ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,8,11,14-Eicosatrienoic Acid ,0302 clinical medicine ,medicine ,Animals ,Premovement neuronal activity ,030304 developmental biology ,Epoxide Hydrolases ,Neurons ,0303 health sciences ,Chemistry ,Original Articles ,Blood flow ,Electric Stimulation ,Capillaries ,Cell biology ,Mice, Inbred C57BL ,Arterioles ,Microscopy, Fluorescence, Multiphoton ,medicine.anatomical_structure ,Neurology ,Regional Blood Flow ,Vasoconstriction ,Microangiography ,cardiovascular system ,Neurology (clinical) ,Pericyte ,Pericytes ,Cardiology and Cardiovascular Medicine ,Flux (metabolism) ,Tomography, Optical Coherence ,030217 neurology & neurosurgery - Abstract
Local blood flow in the brain is tightly coupled to metabolic demands, a phenomenon termed functional hyperemia. Both capillaries and arterioles contribute to the hyperemic response to neuronal activity via different mechanisms and timescales. The nature and specific signaling involved in the hyperemic response of capillaries versus arterioles, and their temporal relationship are not fully defined. We determined the time-dependent changes in capillary flux and diameter versus arteriolar velocity and flow following whisker stimulation using optical microangiography (OMAG) and two-photon microscopy. We further characterized depth-resolved responses of individual capillaries versus capillary networks. We hypothesized that capillaries respond first to neuronal activation, and that they exhibit a coordinated response mediated via endothelial-derived epoxyeicosatrienoates (EETs) acting on pericytes. To visualize peri-capillary pericytes, we used Tie2-GFP/NG2-DsRed mice, and to determine the role of endothelial-derived EETs, we compared cerebrovascular responses to whisker stimulation between wild-type mice and mice with lower endothelial EETs (Tie2-hsEH). We found that capillaries respond immediately to neuronal activation in an orchestrated network-level manner, a response attenuated in Tie2-hsEH and inhibited by blocking EETs action on pericytes. These results demonstrate that capillaries are first responders during functional hyperemia, and that they exhibit a network-level response mediated via endothelial-derived EETs’ action on peri-capillary pericytes.
- Published
- 2021
- Full Text
- View/download PDF
41. Author response: Aquaporin-4-dependent glymphatic solute transport in the rodent brain
- Author
-
Matthew J. Simon, Ming Xiao, Virginia Plá, Wenyan Zou, Humberto Mestre, Tinglin Pu, Alexander S. Thrane, Xiaowen Wang, Ruth M. Castellanos Rivera, Weixi Feng, Masato Yasui, Hiromu Monai, Anna Lr Xavier, Hajime Hirase, Jeffrey J. Iliff, Benjamin A. Plog, Giridhar Murlidharan, Benjamin T. Kress, Yoichiro Abe, John H. Thomas, Ting Du, Lauren M. Hablitz, Iben Lundgaard, Aravind Asokan, and Martin M. Pike
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,Aquaporin 4 ,Rodent ,biology ,Chemistry ,biology.animal ,Glymphatic system ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
42. Analysis of convective and diffusive transport in the brain interstitium
- Author
-
Jeffrey J. Iliff, Lori Ray, and Jeffrey J. Heys
- Subjects
0301 basic medicine ,Convection ,Work (thermodynamics) ,Convective flow ,Extracellular transport ,Flow (psychology) ,Finite Element Analysis ,Models, Neurological ,Brain tissue ,Biotransport ,lcsh:RC346-429 ,Diffusion ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Animals ,Computer Simulation ,lcsh:Neurology. Diseases of the nervous system ,Bulk flow ,Physics ,Research ,Brain ,Biological Transport ,General Medicine ,Mechanics ,Convective velocity ,030104 developmental biology ,Neurology ,Parenchyma ,Hydrodynamics ,Current (fluid) ,Real time iontophoresis ,030217 neurology & neurosurgery ,Algorithms ,Finite element model - Abstract
Background Despite advances in in vivo imaging and experimental techniques, the nature of transport mechanisms in the brain remain elusive. Mathematical modelling verified using available experimental data offers a powerful tool for investigating hypotheses regarding extracellular transport of molecules in brain tissue. Here we describe a tool developed to aid in investigation of interstitial transport mechanisms, especially the potential for convection (or bulk flow) and its relevance to interstitial solute transport, for which there is conflicting evidence. Methods In this work, we compare a large body of published experimental data for transport in the brain to simulations of purely diffusive transport and simulations of combined convective and diffusive transport in the brain interstitium, incorporating current theories of perivascular influx and efflux. Results The simulations show (1) convective flow in the interstitium potentially of a similar magnitude to diffusive transport for molecules of interest and (2) exchange between the interstitium and perivascular space, whereby fluid and solutes may enter or exit the interstitium, are consistent with the experimental data. Simulations provide an upper limit for superficial convective velocity magnitude (approximately \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$v$$\end{document}v = 50 μm min−1), a useful finding for researchers developing techniques to measure interstitial bulk flow. Conclusions For the large molecules of interest in neuropathology, bulk flow may be an important mechanism of interstitial transport. Further work is warranted to investigate the potential for bulk flow. Electronic supplementary material The online version of this article (10.1186/s12987-019-0126-9) contains supplementary material, which is available to authorized users.
- Published
- 2018
43. Transcriptional network analysis of human astrocytic endfoot genes reveals region-specific associations with dementia status and tau pathology
- Author
-
Erin L. Boespflug, Jeffrey J. Iliff, Natalie E. Roese, Charles Murchison, Randall L. Woltjer, Marie Xun Wang, and Matthew J. Simon
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Amyloid beta ,lcsh:Medicine ,Models, Biological ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Western blot ,Parietal Lobe ,Dystrobrevin ,Dystroglycan ,medicine ,Humans ,Dementia ,lcsh:Science ,Aged ,Aged, 80 and over ,Aquaporin 4 ,Temporal cortex ,Amyloid beta-Peptides ,Multidisciplinary ,biology ,medicine.diagnostic_test ,Gene Expression Profiling ,lcsh:R ,medicine.disease ,030104 developmental biology ,Gene Expression Regulation ,Tauopathies ,Organ Specificity ,Astrocytes ,biology.protein ,Female ,lcsh:Q ,Glymphatic system ,030217 neurology & neurosurgery - Abstract
The deposition of misfolded proteins, including amyloid beta plaques and neurofibrillary tangles is the histopathological hallmark of Alzheimer’s disease (AD). The glymphatic system, a brain-wide network of perivascular pathways that supports interstitial solute clearance, is dependent upon expression of the perivascular astroglial water channel aquaporin-4 (AQP4). Impairment of glymphatic function in the aging rodent brain is associated with reduced perivascular AQP4 localization, and in human subjects, reduced perivascular AQP4 localization is associated with AD diagnosis and pathology. Using human transcriptomic data, we demonstrate that expression of perivascular astroglial gene products dystroglycan (DAG1), dystrobrevin (DTNA) and alpha-syntrophin (SNTA1), are associated with dementia status and phosphorylated tau (P-tau) levels in temporal cortex. Gene correlation analysis reveals altered expression of a cluster of potential astrocytic endfoot components in human subjects with dementia, with increased expression associated with temporal cortical P-tau levels. The association between perivascular astroglial gene products, including DTNA and megalencephalic leukoencephalopathy with subcortical cysts 1 (MLC1) with AD status was confirmed in a second human transcriptomic dataset and in human autopsy tissue by Western blot. This suggests changes in the astroglial endfoot domain may underlie vulnerability to protein aggregation in AD.
- Published
- 2018
- Full Text
- View/download PDF
44. Characterization of dural sinus-associated lymphatic vasculature in human Alzheimer’s dementia subjects
- Author
-
Jeffrey J. Iliff, Amanda W. Lund, Zachariah O. Adham, James R. Goodman, and Randall L. Woltjer
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Meningeal lymphatic vessels ,Amyloid beta ,Immunology ,Central nervous system ,Article ,03 medical and health sciences ,Behavioral Neuroscience ,0302 clinical medicine ,Cerebrospinal fluid ,Meninges ,Dural sinus ,Alzheimer Disease ,Parenchyma ,medicine ,Humans ,Aged ,Lymphatic Vessels ,Aged, 80 and over ,Amyloid beta-Peptides ,Membrane Glycoproteins ,Microscopy, Confocal ,biology ,Endocrine and Autonomic Systems ,business.industry ,Brain ,Middle Aged ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,biology.protein ,Female ,Nervous System Diseases ,business ,Glymphatic System ,030217 neurology & neurosurgery - Abstract
Recent reports describing lymphatic vasculature in the meninges have challenged the traditional understanding of interstitial solute clearance from the central nervous system, although the significance of this finding in human neurological disease remains unclear. To begin to define the role of meningeal lymphatic function in the clearance of interstitial amyloid beta (Aβ), and the contribution that its failure may make to the development of Alzheimer's disease (AD), we examined meningeal tissue from a case series including AD and control subjects by confocal microscopy. Our findings confirm the presence of lymphatic vasculature in the human meninges and indicate that, unlike perivascular efflux pathways in the brain parenchyma in subjects with AD, Aβ is not deposited in or around meningeal lymphatic vessels associated with dural sinuses. Our findings demonstrate that while the meningeal lymphatic vasculature may serve as an efflux route for Aβ from the brain and cerebrospinal fluid, Aβ does not deposit in the walls of meningeal lymphatic vessels in the setting of AD.
- Published
- 2018
45. Aquaporin-4-dependent glymphatic solute transport in the rodent brain
- Author
-
Wenyan Zou, Benjamin A. Plog, Ruth M. Castellanos Rivera, Benjamin T. Kress, Iben Lundgaard, Tinglin Pu, Yoichiro Abe, Aravind Asokan, Xiaowen Wang, Anna L.R. Xavier, Matthew J. Simon, Masato Yasui, Ting Du, Humberto Mestre, Hajime Hirase, Alexander S. Thrane, Hiromu Monai, Giridhar Murlidharan, Lauren M. Hablitz, Ming Xiao, Virginia Plá, Martin M. Pike, John H. Thomas, Weixi Feng, and Jeffrey J. Iliff
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Rodent ,Mouse ,QH301-705.5 ,Science ,aquaporin-4 ,General Biochemistry, Genetics and Molecular Biology ,cerebrospinal fluid ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Interstitial fluid ,biology.animal ,Internal medicine ,medicine ,Animals ,Biology (General) ,replication study ,Aquaporin 4 ,Mice, Knockout ,General Immunology and Microbiology ,biology ,General Neuroscience ,glymphatic ,Brain ,Biological Transport ,Extracellular Fluid ,General Medicine ,Anatomy ,Rats ,meta-analysis ,030104 developmental biology ,Endocrinology ,Water channel ,Astrocytes ,solute transport ,Medicine ,Glymphatic system ,sense organs ,Glymphatic System ,030217 neurology & neurosurgery ,Research Article ,Neuroscience - Abstract
The glymphatic system is a brain-wide clearance pathway; its impairment contributes to the accumulation of amyloid-β. Influx of cerebrospinal fluid (CSF) depends upon the expression and perivascular localization of the astroglial water channel aquaporin-4 (AQP4). Prompted by a recent failure to find an effect of Aqp4 knock-out (KO) on CSF and interstitial fluid (ISF) tracer transport, five groups re-examined the importance of AQP4 in glymphatic transport. We concur that CSF influx is higher in wild-type mice than in four different Aqp4 KO lines and in one line that lacks perivascular AQP4 (Snta1 KO). Meta-analysis of all studies demonstrated a significant decrease in tracer transport in KO mice and rats compared to controls. Meta-regression indicated that anesthesia, age, and tracer delivery explain the opposing results. We also report that intrastriatal injections suppress glymphatic function. This validates the role of AQP4 and shows that glymphatic studies must avoid the use of invasive procedures.
- Published
- 2018
- Full Text
- View/download PDF
46. Abstract WP411: Identification and Characterization of Human Meningeal Lymphatics
- Author
-
Randall L. Woltjer, James R. Goodman, Zachariah O. Adham, and Jeffrey J. Iliff
- Subjects
Advanced and Specialized Nursing ,Pathology ,medicine.medical_specialty ,business.industry ,Meningeal lymphatic vessels ,Dura mater ,Central nervous system ,Meninges ,medicine.disease ,medicine.anatomical_structure ,Lymphatic system ,Cerebrospinal fluid ,medicine ,Dementia ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,Superior sagittal sinus - Abstract
Late onset Alzheimer’s Disease (AD) is the most common form of dementia, affecting 40 million patients and countless families worldwide. AD is characterized by accumulation of amyloid beta (Aß) in the brain and central nervous system (CNS). A growing body of evidence indicates that while Aß production is unchanged, its clearance from the CNS is attenuated in late onset AD. Therefore, it is critical to determine how Aβ is cleared from the brain and if these processes differ in healthy and AD patients. Recently, two groups independently reported that a network of meningeal lymphatic vasculature participates in the clearance of solutes and macromolecules from the brain and cerebrospinal fluid of mice. However the presence of meningeal lymphatic vessels and their potential role in the clearance of Aß has not yet been defined in humans. Therefore, to determine if human meningeal lymphatic vessels absorb Aβ, we used immunofluorescence to examine superior sagittal sinus-associated dura mater tissue in a cohort of 21 patient samples including 6 subjects with no diagnosed dementia (control), 7 subjects with histopathologically confirmed AD, and 8 with diagnosis of mixed or other dementia. We found podoplanin (PDPN) positive, lumenized vessels in 19/21 patient samples, with 5/6 in control patients, 7/7 in AD patients and 7/8 in other or mixed dementia patients. These vessels were located in the dura mater, lateral to the superior sagittal sinus, and ranged from approximately 10 to 500 microns in diameter. Trace Aβ immunoreactivity was colocalized with podoplanin-positive vessels in 1/5 control, 1/7 AD, and 1/7 other or mixed dementia patients. Aß reactivity was found in meningeal blood vessels of 0/6 control, 1/7 AD, and 0/7 other or mixed dementia patients. To our knowledge, these data are the first to robustly support the existence of lymphatic vasculature in the human meninges. The data further suggest that Aß does not appear to widely deposit along wither these meningeal lymphatic or blood vessels, even among AD subjects.
- Published
- 2018
- Full Text
- View/download PDF
47. P3-189: CHARACTERISING THE ASTROCYTIC PROFILE OF DYSTROBREVIN IN THE BRAIN DURING ALZHEIMER'S DISEASE
- Author
-
Ozama Ismail, Marie Xun Wang, and Jeffrey J. Iliff
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Dystrobrevin ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,Biology ,Neuroscience - Published
- 2019
- Full Text
- View/download PDF
48. Methamphetamine induces the release of endothelin
- Author
-
Jeong Woo Seo, G. Alexander West, Susan M. Jones, Jeffrey J. Iliff, and Trisha A. Hostetter
- Subjects
Endothelium ,business.industry ,Endothelin receptor antagonist ,Neurotoxicity ,030204 cardiovascular system & hematology ,Pharmacology ,Methamphetamine ,medicine.disease ,Endothelin 1 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine.anatomical_structure ,Dopamine ,medicine ,medicine.symptom ,business ,Endothelin receptor ,030217 neurology & neurosurgery ,Vasoconstriction ,medicine.drug - Abstract
Methamphetamine is a potent psychostimulant drug of abuse that increases release and blocks reuptake of dopamine, producing intense euphoria, factors that may contribute to its widespread abuse. It also produces severe neurotoxicity resulting from oxidative stress, DNA damage, blood-brain barrier disruption, microgliosis, and mitochondrial dysfunction. Intracerebral hemorrhagic and ischemic stroke have been reported after intravenous and oral abuse of methamphetamine. Several studies have shown that methamphetamine causes vasoconstriction of vessels. This study investigates the effect of methamphetamine on endothelin-1 (ET-1) release in mouse brain endothelial cells by ELISA. ET-1 transcription as well as endothelial nitric oxide synthase (eNOS) activation and transcription were measured following methamphetamine treatment. We also examine the effect of methamphetamine on isolated cerebral arteriolar vessels from C57BL/6 mice. Penetrating middle cerebral arterioles were cannulated at both ends with a micropipette system. Methamphetamine was applied extraluminally, and the vascular response was investigated. Methamphetamine treatment of mouse brain endothelial cells resulted in ET-1 release and a transient increase in ET-1 message. The activity and transcription of eNOS were only slightly enhanced after 24 hr of treatment with methamphetamine. In addition, methamphetamine caused significant vasoconstriction of isolated mouse intracerebral arterioles. The vasoconstrictive effect of methamphetamine was attenuated by coapplication of the endothelin receptor antagonist PD145065. These findings suggest that vasoconstriction induced by methamphetamine is mediated through the endothelin receptor and may involve an endothelin-dependent pathway.
- Published
- 2015
- Full Text
- View/download PDF
49. [IC‐P‐157]: FULLY AUTOMATED, WHOLE BRAIN MORPHOLOGICAL SEGMENTATION OF ENLARGED PERIVASCULAR SPACES AT CLINICAL FIELD STRENGTH: MRI‐BASED MULTIMODAL AUTOIDENTIFICATION OF PERIVASCULAR SPACES (MMAPS)
- Author
-
Jeffrey Kaye, Erin L. Boespflug, Lisa C. Silbert, Jeffrey M. Pollock, William D. Rooney, Jeffrey J. Iliff, Daniel Schwartz, and David Lahna
- Subjects
Epidemiology ,Computer science ,Health Policy ,Field strength ,Anatomy ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Developmental Neuroscience ,Fully automated ,medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,Perivascular space ,Morphological segmentation ,Neuroscience - Published
- 2017
- Full Text
- View/download PDF
50. A transcriptome-based assessment of the astrocytic dystrophin associated complex in the developing human brain
- Author
-
Matthew J. Simon, Charles Murchison, and Jeffrey J. Iliff
- Subjects
Adult ,Male ,0301 basic medicine ,Candidate gene ,Adolescent ,Central nervous system ,Muscle Proteins ,Article ,Dystrophin-Associated Protein Complex ,Transcriptome ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Gene expression ,medicine ,Humans ,Child ,Gene ,Aquaporin 4 ,Analysis of Variance ,biology ,Calcium-Binding Proteins ,Neuropeptides ,Brain ,Membrane Proteins ,Human brain ,Gene Ontology ,030104 developmental biology ,medicine.anatomical_structure ,Astrocytes ,Dystrophin-Associated Proteins ,biology.protein ,Female ,Glymphatic system ,Dystrophin ,Neuroscience ,030217 neurology & neurosurgery ,Subcellular Fractions - Abstract
Astrocytes play a critical role in regulating the interface between the cerebral vasculature and the central nervous system. Contributing to this is the astrocytic endfoot domain, a specialized structure that ensheathes the entirety of the vasculature and mediates signaling between endothelial cells, pericytes and neurons. The astrocytic endfoot has been implicated as a critical element of the glymphatic pathway and changes in protein expression profiles in this cellular domain are linked to Alzheimer’s disease pathology. Despite this, basic physiological properties of this structure remain poorly understood including the developmental timing of its formation, and the protein components that localize there to mediate its functions. Here we use human transcriptome data from male and female subjects across several developmental stages and brain regions to characterize the gene expression profile of the dystrophin associated complex (DAC), a known structural component of the astrocytic endfoot that supports perivascular localization of the astroglial water channel aquaporin-4 (AQP4). Transcriptomic profiling is also used to define genes exhibiting parallel expression profiles to DAC elements, generating a pool of candidate genes that encode gene products that may contribute to the physiological function of the perivascular astrocytic endfoot domain. We found that several genes encoding transporter proteins are transcriptionally associated with DAC genes.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.