Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters











Publication year range
1.
Vitam Horm ; 126: 77-96, 2024.
Article in English | MEDLINE | ID: mdl-39029977

ABSTRACT

Sleep is a physiological process that preserves the integrity of the neuro-immune-endocrine network to maintain homeostasis. Sleep regulates the production and secretion of hormones, neurotransmitters, cytokines and other inflammatory mediators, both at the central nervous system (CNS) and at the periphery. Sleep promotes the removal of potentially toxic metabolites out of the brain through specialized systems such as the glymphatic system, as well as the expression of specific transporters in the blood-brain barrier. The blood-brain barrier maintains CNS homeostasis by selectively transporting metabolic substrates and nutrients into the brain, by regulating the efflux of metabolic waste products, and maintaining bidirectional communication between the periphery and the CNS. All those processes are disrupted during sleep loss. Brain endothelial cells express the blood-brain barrier phenotype, which arises after cell-to-cell interactions with mural cells, like pericytes, and after the release of soluble factors by astroglial endfeet. Astroglia, pericytes and brain endothelial cells respond differently to sleep loss; evidence has shown that sleep loss induces a chronic low-grade inflammatory state at the CNS, which is associated with blood-brain barrier dysfunction. In animal models, blood-brain barrier dysfunction is characterized by increased blood-brain barrier permeability, decreased tight junction protein expression and pericyte detachment from the capillary wall. Blood-brain barrier dysfunction may promote defects in brain clearance of potentially neurotoxic metabolites and byproducts of neural physiology, which may eventually contribute to neurodegenerative diseases. This chapter aims to describe the cellular and molecular mechanisms by which sleep loss modifies the function of the blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Sleep Deprivation , Blood-Brain Barrier/metabolism , Humans , Animals , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Endothelial Cells/metabolism
2.
Adv Clin Chem ; 121: 1-88, 2024.
Article in English | MEDLINE | ID: mdl-38797540

ABSTRACT

The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.


Subject(s)
Biomarkers , Blood-Brain Barrier , Blood-Brain Barrier/metabolism , Humans , Biomarkers/metabolism , Animals
3.
mBio ; 15(3): e0325223, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38289074

ABSTRACT

Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE: A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.


Subject(s)
Bacterial Infections , Pericytes , Animals , Mice , Sheep , Pericytes/metabolism , Endothelial Cells , Connexin 43/metabolism , Connexin 43/pharmacology , Inflammation , Bacterial Infections/metabolism
4.
Braz J Cardiovasc Surg ; 38(6): e20230045, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37797088

ABSTRACT

This short article discusses selected scanning electron microscope and transmission electron microscope features of vasa vasorum including pericytes and basement membrane of the human saphenous vein (SV) harvested with either conventional (CON) or no-touch (NT) technique for coronary artery bypass grafting. Scanning electron microscope data shows the general damage to vasa vasorum of CON-SV, while the transmission electron microscope data presents ultrastructural features of the vasa in more detail. Hence there are some features suggesting pericyte involvement in the contraction of vasa blood vessels, particularly in CON-SV. Other features associated with the vasa vasorum of both CON-SV and NT-SV preparations include thickened and/or multiplied layers of the basement membrane. In some cases, multiple layers of basement membrane embrace both pericyte and vasa microvessel making an impression of a "unit" made by basement membrane-pericyte-endothelium/microvessel. It can be speculated that this structural arrangement has an effect on the contractile and/or relaxing properties of the vessels involved. Endothelial colocalization of immunoreactive inducible nitric oxide synthase and endothelin-1 can be observed (with laser confocal microscope) in some of the vasa microvessels. It can be speculated that this phenomenon, particularly of the expression of inducible nitric oxide synthase, might be related to structurally changed vasa vessels, e.g., with expanded basement membrane. Fine physiological relationships between vasa vasorum endothelium, basement membrane, pericyte, and perivascular nerves have yet to be uncovered in the detail needed for better understanding of the cells'specific effects in SV preparations for coronary artery bypass grafting.


Subject(s)
Saphenous Vein , Vasa Vasorum , Humans , Saphenous Vein/transplantation , Nitric Oxide Synthase Type II/metabolism , Vasa Vasorum/metabolism , Vasa Vasorum/ultrastructure , Coronary Artery Bypass/methods , Endothelium, Vascular
5.
Int J Mol Sci ; 24(13)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37446312

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Histopathologically, AD presents two pathognomonic hallmarks: (1) neurofibrillary tangles, characterized by intracellular deposits of hyperphosphorylated tau protein, and (2) extracellular amyloid deposits (amyloid plaques) in the brain vasculature (cerebral amyloid angiopathy; CAA). It has been proposed that vascular amyloid deposits could trigger neurovascular unit (NVU) dysfunction in AD. The NVU is composed primarily of astrocytic feet, endothelial cells, pericytes, and basement membrane. Although physical exercise is hypothesized to have beneficial effects against AD, it is unknown whether its positive effects extend to ameliorating CAA and improving the physiology of the NVU. We used the triple transgenic animal model for AD (3xTg-AD) at 13 months old and analyzed through behavioral and histological assays, the effect of voluntary physical exercise on cognitive functions, amyloid angiopathy, and the NVU. Our results show that 3xTg-AD mice develop vascular amyloid deposits which correlate with cognitive deficits and NVU alteration. Interestingly, the physical exercise regimen decreases amyloid angiopathy and correlates with an improvement in cognitive function as well as in the underlying integrity of the NVU components. Physical exercise could represent a key therapeutic approach in cerebral amyloid angiopathy and NVU stability in AD patients.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Mice , Animals , Alzheimer Disease/metabolism , Plaque, Amyloid/metabolism , Endothelial Cells/metabolism , Mice, Transgenic , Cerebral Amyloid Angiopathy/metabolism , Brain/metabolism , Disease Models, Animal , Amyloid beta-Peptides/metabolism
6.
Front Cell Dev Biol ; 11: 1148121, 2023.
Article in English | MEDLINE | ID: mdl-36936686

ABSTRACT

Cultured mesenchymal stromal cells are among the most used cells in clinical trials. Currently, their potential benefits include provision of mature cell types through differentiation, and secretion of various types of paracrine signaling molecules. Even though research on these cells has spanned some decades now, surprisingly, their therapeutic potential has not been fully translated into clinical practice yet, which calls for further understanding of their intrinsic nature and modes of action. In this review, after discussing pieces of evidence that suggest that some perivascular cells may exhibit mesenchymal stem cell characteristics in vivo, we examine the possibility that subpopulations of perivascular and/or adventitial cells activated after tissue injury behave as MSCs and contribute to the resolution of tissue injury by providing cues for the development of regenerative macrophages at injured sites. Under this perspective, an important contribution of cultured MSCs (or their acellular products, such as extracellular vesicles) used in cell therapies would be to instigate the development of M2-like macrophages that support the tissue repair process.

7.
Cell Mol Neurobiol ; 43(2): 525-541, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35195811

ABSTRACT

Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.


Subject(s)
Neuroinflammatory Diseases , Pericytes , Adult , Humans , Brain/blood supply , Blood-Brain Barrier , Central Nervous System
8.
Rev. bras. cir. cardiovasc ; Rev. bras. cir. cardiovasc;38(6): e20230045, 2023. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1507837

ABSTRACT

ABSTRACT This short article discusses selected scanning electron microscope and transmission electron microscope features of vasa vasorum including pericytes and basement membrane of the human saphenous vein (SV) harvested with either conventional (CON) or no-touch (NT) technique for coronary artery bypass grafting. Scanning electron microscope data shows the general damage to vasa vasorum of CON-SV, while the transmission electron microscope data presents ultrastructural features of the vasa in more detail. Hence there are some features suggesting pericyte involvement in the contraction of vasa blood vessels, particularly in CON-SV. Other features associated with the vasa vasorum of both CON-SV and NT-SV preparations include thickened and/or multiplied layers of the basement membrane. In some cases, multiple layers of basement membrane embrace both pericyte and vasa microvessel making an impression of a "unit" made by basement membrane-pericyte-endothelium/microvessel. It can be speculated that this structural arrangement has an effect on the contractile and/or relaxing properties of the vessels involved. Endothelial colocalization of immunoreactive inducible nitric oxide synthase and endothelin-1 can be observed (with laser confocal microscope) in some of the vasa microvessels. It can be speculated that this phenomenon, particularly of the expression of inducible nitric oxide synthase, might be related to structurally changed vasa vessels, e.g., with expanded basement membrane. Fine physiological relationships between vasa vasorum endothelium, basement membrane, pericyte, and perivascular nerves have yet to be uncovered in the detail needed for better understanding of the cells'specific effects in SV preparations for coronary artery bypass grafting.

9.
Mol Neurobiol ; 59(6): 3738-3754, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35381889

ABSTRACT

Iron deficiency anemia is a prevalent health problem among pregnant women and infants, particularly in the developing countries that causes brain development deficits and poor cognitive outcomes. Since tissue iron depletion may impair myelination and trigger cellular hypoxic signaling affecting blood vessels, we studied myelination and the neurovascular unit (NVU) in infant rats born to mothers fed with an iron deficient (ID) or control diet from embryonic day 5 till weaning. Blood samples and brains of rat pups at postnatal day (PND) 14 and 30 were analyzed. PND 14 ID rats had severe microcytic hypochromic anemia that was almost reversed at PND 30 although hypomyelination and astrocyte immature phenotype in the corpus callosum were significant at that age. In CA1 hippocampal region, PND 14 and PND 30 ID rats showed significant reduced expression of the receptor ß of the platelet-derived growth factor localized in pericytes and associated to aquaporin 4 (AQP4) immunopositive capillaries. Shorter AQP4 + capillaries and reduced AQP4 expression were also evidenced in PND 14 and PND 30 ID rats. In addition, pericyte membrane permeability through large-pore channels was transiently increased in ID rats at PND 14 but not at PND 30, while the blood-brain barrier permeability was not affected. Remarkably, transient increased pericyte permeability found in PND 14 ID rats was not directly related to iron depletion, suggesting the involvement of other iron deficiency anemia-induced mechanisms. In summary, severe ID during gestation and lactation produces persistent hypomyelination and significantly affects hippocampal pericytes and astrocytes in the NVU which may trigger impaired neurovascular function.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/metabolism , Animals , Animals, Newborn , Female , Hippocampus/metabolism , Humans , Iron/metabolism , Lactation , Pregnancy , Rats
10.
J Cell Physiol ; 237(4): 2198-2210, 2022 04.
Article in English | MEDLINE | ID: mdl-35040139

ABSTRACT

Pericytes and glial cells are known to collaborate in dental pulp tissue repair. Cell-based therapies that stimulate these stromal components may be of therapeutic relevance for partially vital dental pulp conditions. This study aimed to examine the early effect of photobiomodulation (PBM) in pericytes from experimentally injured pulp tissue. To accomplish this, we used the Nestin-GFP/NG2-DsRed mice, which could allow the identification of distinct pericyte phenotypes. We discovered the presence of two pericytes subsets within the dental pulp, the Nestin + NG2+ (type-2) and Nestin- NG2+ (type-1). Upon injury, PBM treatment led to a significant increase in Nestin+ cells and pericytes. This boost was mainly conferred by the more committed pericyte subset (NestinNG2+ ). PBM also stimulated terminal blood vessels sprouting adjacent to the injury site while maintaining signs of pulp vitality. In vitro, PBM induced VEGF upregulation, improved dental pulp cells proliferation and migration, and favored their mineralization potential. Herein, different subsets of perivascular cells were unveiled in the pulp tissue. PBM enhanced not only NG2+ cells but nestin-expressing progenitors in the injured dental pulp.


Subject(s)
Dental Pulp/cytology , Neuroglia , Pericytes , Animals , Mice , Nestin/genetics , Transgenes
11.
Cell Mol Neurobiol ; 42(3): 557-564, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33010018

ABSTRACT

Glioma is the prevalent aggressive primary brain tumor, with a very poor prognosis. The absence of advanced understanding of the roles played by the cells within the glioma microenvironment limits the development of effective drugs. A recent study indicates that periostin expressed by pericytes is crucial for glioma angiogenesis. Here, we describe succinctly the results and implications of this discovery in what we know about pericytes within the glioma microenvironment. The emerging knowledge from this work will benefit the development of therapies for gliomas.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/pathology , Glioma/pathology , Humans , Morphogenesis , Neovascularization, Pathologic/pathology , Pericytes/pathology , Tumor Microenvironment
12.
Neurotox Res ; 39(6): 2154-2174, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34677787

ABSTRACT

The blood-brain barrier is a dynamic structure, collectively referred to as the neurovascular unit. It is responsible for the exchange of blood, oxygen, ions, and other molecules between the peripheral circulation and the brain compartment. It is the main entrance to the central nervous system and as such critical for the maintenance of its homeostasis. Dysfunction of the blood-brain barrier is a characteristic of several neurovascular pathologies. Moreover, physiological changes, environmental factors, nutritional habits, and psychological stress can modulate the tightness of the barrier. In this contribution, we summarize our current understanding of structure and function of this important component of the brain. We also describe the neurological deficits associated with its damage. A special emphasis is placed in the effect of the exposure to xenobiotics and pollutants in the permeability of the barrier. Finally, current protective strategies as well as the culture models to study this fascinating structure are discussed.


Subject(s)
Blood-Brain Barrier/physiology , Brain/physiology , Animals , Blood-Brain Barrier/anatomy & histology , Blood-Brain Barrier/metabolism , Brain/anatomy & histology , Brain/metabolism , Humans
13.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188608, 2021 12.
Article in English | MEDLINE | ID: mdl-34384850

ABSTRACT

Cancer cells are embedded within the tumor microenvironment and interact dynamically with its components during tumor progression. Understanding the molecular mechanisms by which the tumor microenvironment components communicate is crucial for the success of therapeutic applications. Recent studies show, by using state-of-the-art technologies, including sophisticated in vivo inducible Cre/loxP mediated systems and CRISPR-Cas9 gene editing, that pericytes communicate with cancer cells. The arising knowledge on cross-talks within the tumor microenvironment will be essential for the development of new therapies against cancer. Here, we review recent progress in our understanding of pericytes roles within tumors.


Subject(s)
Pericytes/metabolism , Tumor Microenvironment/immunology , Humans
14.
Int J Mol Sci ; 22(15)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34361013

ABSTRACT

Glioblastoma (GBM) is the most aggressive and common primary tumor of the central nervous system. It is characterized by having an infiltrating growth and by the presence of an excessive and aberrant vasculature. Some of the mechanisms that promote this neovascularization are angiogenesis and the transdifferentiation of tumor cells into endothelial cells or pericytes. In all these processes, the release of extracellular microvesicles by tumor cells plays an important role. Tumor cell-derived extracellular microvesicles contain pro-angiogenic molecules such as VEGF, which promote the formation of blood vessels and the recruitment of pericytes that reinforce these structures. The present study summarizes and discusses recent data from different investigations suggesting that Netrin-1, a highly versatile protein recently postulated as a non-canonical angiogenic ligand, could participate in the promotion of neovascularization processes in GBM. The relevance of determining the angiogenic signaling pathways associated with the interaction of Netrin-1 with its receptors is posed. Furthermore, we speculate that this molecule could form part of the microvesicles that favor abnormal tumor vasculature. Based on the studies presented, this review proposes Netrin-1 as a novel biomarker for GBM progression and vascularization.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neovascularization, Pathologic/genetics , Netrin-1/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Neovascularization, Pathologic/metabolism , Netrin Receptors/genetics , Netrin Receptors/metabolism , Netrin-1/genetics , Signal Transduction
15.
Autops Case Rep ; 11: e2021262, 2021.
Article in English | MEDLINE | ID: mdl-34307223

ABSTRACT

We performed autopsies on two cases of COVID-19. The microcirculations of all organs were the site of the pathological findings. Thrombotic microangiopathy was found in the brain and also the kidneys. Vasculitis was also a feature of the autopsy findings, together with portal triaditis of the liver. The major pathological findings in both cases were fibrin deposits. Within the lung, the fibrin deposits were observed in the alveolar microcirculation in sub-endothelial locations of capillaries, arterioles, post capillary venules, and the adventitia of larger vessels. These fibrin deposits in the lungs occurred at the sites where pericytes are located in these vessels. The pericyte with its high concentration of ACE-2 receptors and its procoagulant state may represent one of the primary sites of action of SARS-CoV-2. A review of pericytes in health and disease is undertaken. COVID-19 is a disease of the microcirculation.

16.
Stem Cell Res Ther ; 12(1): 183, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33726849

ABSTRACT

BACKGROUND: Peripheral arterial disease (PAD) affects millions of people and compromises quality of life. Critical limb ischemia (CLI), which is the most advanced stage of PAD, can cause nonhealing ulcers and strong chronic pain, and it shortens the patients' life expectancy. Cell-based angiogenic therapies are becoming a real therapeutic approach to treat CLI. Pericytes are cells that surround vascular endothelial cells to reinforce vessel integrity and regulate local blood pressure and metabolism. In the past decade, researchers also found that pericytes may function as stem or progenitor cells in the body, showing the potential to differentiate into several cell types. We investigated the gene expression profiles of pericytes during the early stages of limb ischemia, as well as the alterations in pericyte subpopulations to better understand the behavior of pericytes under ischemic conditions. METHODS: In this study, we used a hindlimb ischemia model to mimic CLI in C57/BL6 mice and explore the role of pericytes in regeneration. To this end, muscle pericytes were isolated at different time points after the induction of ischemia. The phenotypes and transcriptomic profiles of the pericytes isolated at these discrete time points were assessed using flow cytometry and RNA sequencing. RESULTS: Ischemia triggered proliferation and migration and upregulated the expression of myogenesis-related transcripts in pericytes. Furthermore, the transcriptomic analysis also revealed that pericytes induce or upregulate the expression of a number of cytokines with effects on endothelial cells, leukocyte chemoattraction, or the activation of inflammatory cells. CONCLUSIONS: Our findings provide a database that will improve our understanding of skeletal muscle pericyte biology under ischemic conditions, which may be useful for the development of novel pericyte-based cell and gene therapies.


Subject(s)
Pericytes , Transcriptome , Animals , Chemotaxis, Leukocyte , Endothelial Cells , Humans , Ischemia/genetics , Mice , Muscle, Skeletal , Quality of Life
17.
J Neurochem ; 157(4): 1377-1391, 2021 05.
Article in English | MEDLINE | ID: mdl-32974913

ABSTRACT

Perivascular pericytes are key regulators of the blood-brain barrier, vascular development, and cerebral blood flow. Deciphering pericyte roles in health and disease requires cellular tracking; yet, pericyte identification remains challenging. A previous study reported that the far-red fluorophore TO-PRO-3 (642/661), usually employed as a nuclear dye in fixed tissue, was selectively captured by live pericytes from the subventricular zone. Herein, we validated TO-PRO-3 as a specific pericyte tracer in the nervous system (NS). Living pericytes from ex vivo murine hippocampus, cortex, spinal cord, and retina robustly incorporated TO-PRO-3. Classical pericyte immunomarkers such as chondroitin sulphate proteoglycan neuron-glial antigen 2 (NG2) and platelet-derived growth factor receptor beta antigen (PDGFrß) and the new pericyte dye NeuroTrace 500/525 confirmed cellular specificity of dye uptake. The TO-PRO-3 signal enabled quantification of pericytes density and morphometry; likewise, TO-PRO-3 labeling allowed visualization of pericytes associated with other components of the neurovascular unit. A subset of TO-PRO-3 stained cells expressed the contractile protein α-SMA, indicative of their ability to control the capillary diameter. Uptake of TO-PRO-3 was independent of connexin/pannexin channels but was highly sensitive to temperature and showed saturation, suggesting that a yet unidentified protein-mediated active transport sustained dye incorporation. We conclude that TO-PRO-3 labeling provides a reliable and simple tool for the bioimaging of pericytes in the murine NS microvasculature.


Subject(s)
Carbocyanines , Fluorescent Dyes , Pericytes , Staining and Labeling/methods , Animals , Mice
18.
Methods Mol Biol ; 2367: 37-46, 2021.
Article in English | MEDLINE | ID: mdl-32789776

ABSTRACT

The study of the regionalized function of the blood-brain barrier at the level of brain endothelial cells and pericytes is essential to understand the biological properties and molecular mechanisms regulating this biological barrier. The isolation of blood vessels from specific brain regions will allow to understand regional differences in susceptibility to pathological phenomena such as ischemia, traumatic brain injury, and neurodegenerative diseases, such as Alzheimer disease. Here, we propose an efficient and fast method to isolate brain endothelial cells and pericytes from a specific cerebral region. The isolated brain endothelial cells and pericytes are viable to perform conventional molecular and histological techniques such as Western blots, immunocytofluorescence, and scanning electron microscopy.


Subject(s)
Brain , Blood-Brain Barrier , Endothelial Cells , Microvessels , Pericytes
19.
Belo Horizonte; s.n; 2021. 67 p. ilus.
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: biblio-1392378

ABSTRACT

Em indivíduos com diabetes mellitus (DM) o reparo tecidual cutâneo atrasado representa um desafio para o sistema de saúde. Evidências recentes mostram o potencial da fotobiomodulação (PBM, do inglês, photobiomodulation) em induzir a diferenciação de células-tronco em múltiplos tecidos. Os pericitos são células-tronco perivasculares com ampla plasticidade, podendo ser considerados alvos potenciais para a PBM durante o reparo tecidual. Assim, o objetivo deste estudo foi investigar o efeito da PBM na modulação de células indiferenciadas em feridas de camundongos em condição sistêmica análoga ao DM tipo-II. Trata-se de um estudo in vivo (CEUA#62/2019) utilizando camundongos transgênicos diabéticos induzidos artificialmente e com marcação endógena para pericitos (NG2+/DsRed+; Nestina+/GFP+ & NG2+/DsRed+) e células mesenquimais indiferenciadas (Nestina+/GFP+). Foram realizadas bilateralmente feridas no dorso dos camundongos, e as mesmas foram submetidas ou não a PBM e avaliadas nos tempos experimentais 1, 3 e 7 dias. O reparo tecidual foi acompanhado por morfometria, avaliação de fluxo sanguíneo, análises histológicas nos tempos 1, 3 e 7 dias, além de identificação dos pericitos por microscopia confocal ao final de 3 e 7 dias. Os dados obtidos foram submetidos à análise estatística. As análises morfométricas e histológicas mostraram maior efeito de reparo nas feridas submetidas a PBM, onde a média de área remanescente após 1 de PBM foi 73% da medida de área total inicial no grupo PBM e 86,21% no controle (p=0,0257); aos 3 dias, foram 66,98% e 87,49% respectivamente (p=0,026) e aos 7 dias, 25,54% no grupo PBM e 39,43% no controle (p<0,05). A perfusão sanguínea foi maior nas áreas das feridas quando comparadas a pele íntegra, no entanto, não foram observadas diferenças entre as feridas submetidas ou não a PBM. Por outro lado, foram observadas nestas (PBM), maiores quantidades de células mesenquimais indiferenciadas (Nestina+/GFP+) e de pericitos tipo-I (NG2+/DsRed+) após 7 dias. A utilização de PBM em processos de reparo tecidual em modelo diabético de feridas demostraram resultados significativos tanto clínicos com a nível celular, envolvendo em grande parte as células mesenquimais (nestina+/GFP+) e pericitos (NG2+/DsRed+). Conhecer os mecanismos celulares de ação da PBM em feridas de modelo diabético permite controlar esse processo, além de explorar essa técnica e abrir caminhos para investigação de outras ferramentas e protocolos úteis para o tratamento de feridas nestes indivíduos afetados.


In individuals with diabetes mellitus (DM) delayed cutaneous tissue repair represents a challenge for the health system. Recent evidence shows the potential of photobiomodulation (PBM) to induce stem cell differentiation in multiple tissues. Pericytes, in turn, are perivascular stem cells with wide plasticity and can be considered potential targets for PBM during tissue repair. The objective of this study was to investigate the role of PBM in stem cell modulation in wounds of mice under systemic condition analogous to type-II DM. This is an in vivo study (Ethical protocol: CEUA#62/2019) using artificially induced transgenic diabetic mice with endogenous labeling for pericytes (NG2+/DsRed+; Nestin+/GFP+ & NG2+/DsRed+) and undifferentiated mesenchymal cells (Nestin+/GFP+). Wounds on the mice's back were bilaterally performed, and then submitted or not to laser therapy and evaluated at experimental times 1, 3 and 7 days. Tissue repair was followed by periodic measurements of wound diameter, blood flow assessment, histological analysis and screening of pericytes by confocal microscopy at the end of each experimental time. The data obtained were submitted to statistical analysis. The histologic and morphometric analysis showed a greater repair effect in wounds submitted to PBM, where the average area remaining after 1 day of laser application was 73% of the initial total area measurement in the PBM group, and 86.21% in the control (p= 0.0257); at 3 days, they were 66.98% and 87.49% respectively (p= 0. 026), and at 7 days, 25.54% in the PBM group and 39.43% in the control (p<0.05). Blood perfusion was greater in wound areas when compared to intact skin, however, no statistical differences were observed between wounds submitted or not to PBM. On the other hand, larger amounts of undifferentiated mesenchymal cells (Nestin+/GFP+) and type-I pericytes (NG2+/DsRed+) were observed in these wounds after 7 days. The use of PBM in tissue repair processes in a diabetic wound healing model showed significant clinical and cellular results, involving mostly mesenchymal cells (nestin+/GFP+) and pericytes (NG2+/DsRed+). Knowing the cellular mechanisms of action of PBM in wounds of diabetic, allows better management of the therapy, also it opens paths for the investigation of other tools and protocols useful for the treatment of wounds in DM individuals.


Subject(s)
Wound Healing , Pericytes , Diabetes Mellitus , Lasers
20.
Autops. Case Rep ; 11: e2021262, 2021. graf
Article in English | LILACS | ID: biblio-1249006

ABSTRACT

We performed autopsies on two cases of COVID-19. The microcirculations of all organs were the site of the pathological findings. Thrombotic microangiopathy was found in the brain and also the kidneys. Vasculitis was also a feature of the autopsy findings, together with portal triaditis of the liver. The major pathological findings in both cases were fibrin deposits. Within the lung, the fibrin deposits were observed in the alveolar microcirculation in sub-endothelial locations of capillaries, arterioles, post capillary venules, and the adventitia of larger vessels. These fibrin deposits in the lungs occurred at the sites where pericytes are located in these vessels. The pericyte with its high concentration of ACE-2 receptors and its procoagulant state may represent one of the primary sites of action of SARS-CoV-2. A review of pericytes in health and disease is undertaken. COVID-19 is a disease of the microcirculation.


Subject(s)
Humans , Male , Adolescent , Aged, 80 and over , Pericytes , SARS-CoV-2 , Microcirculation , Autopsy
SELECTION OF CITATIONS
SEARCH DETAIL