Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(6): 114288, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38814782

ABSTRACT

Lipids have emerged as potent regulators of immune cell function. In the skin, adipocyte lipolysis increases the local pool of free fatty acids and is essential for coordinating early macrophage inflammation following injury. Here, we investigate G-protein-coupled receptor 84 (GPR84), a medium-chain fatty acid (MCFA) receptor, for its potential to propagate pro-inflammatory signaling after skin injury. GPR84 signaling was identified as a key component of regulating myeloid cell numbers and subsequent tissue repair through in vivo administration of a pharmacological antagonist and the MCFA decanoic acid. We found that impaired injury-induced dermal adipocyte lipolysis is a hallmark of diabetes, and lipidomic analysis demonstrated that MCFAs are significantly reduced in diabetic murine wounds. Furthermore, local administration of decanoic acid rescued myeloid cell numbers and tissue repair during diabetic wound healing. Thus, GPR84 is a readily targetable lipid signaling pathway for manipulating injury-induced tissue inflammation with beneficial effects on acute diabetic healing.


Subject(s)
Diabetes Mellitus, Experimental , Inflammation , Receptors, G-Protein-Coupled , Skin , Wound Healing , Animals , Male , Mice , Adipocytes/metabolism , Decanoic Acids/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Inflammation/pathology , Inflammation/metabolism , Lipolysis/drug effects , Mice, Inbred C57BL , Myeloid Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Skin/pathology , Skin/metabolism , Skin/injuries , Wound Healing/drug effects , Female
2.
Biomedicines ; 9(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34944568

ABSTRACT

Diabetic wound healing is associated with impaired function and reduced numbers of myofibroblasts, a heterogeneous cell population with varying capacities to promote repair. To determine how diabetes alters myofibroblast composition, we performed flow cytometry and spatial tissue analysis of myofibroblast subsets throughout the healing process in diabetic (db/db) and control (db/+) mouse skin. We observed reduced numbers of profibrotic SCA1+; CD34+; CD26+ myofibroblasts in diabetic wounds five days after injury, with decreased expression of fibrosis-associated genes compared to myofibroblasts from db/+ mouse wounds. While the abundance of myofibroblasts remained reduced in db/db mouse wounds compared to controls, the altered myofibroblast heterogeneity and gene expression in diabetic mice was improved seven days after injury. The natural correction of myofibroblast composition and gene expression in db/db wound beds temporally corresponds with a macrophage phenotypic switch. Correlation analysis from individual wound beds revealed that wound healing in control mice is associated with CD206+ macrophages, while the rescued myofibroblast phenotypes in diabetic wounds are correlated with increased CD301b+ macrophage numbers. These data demonstrate how diabetes impacts specific subsets of myofibroblasts and indicate that signaling capable of rescuing impaired diabetic wound healing could be different from signals that regulate wound healing under nonpathological conditions.

3.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669239

ABSTRACT

Irregular inflammatory responses are a major contributor to tissue dysfunction and inefficient repair. Skin has proven to be a powerful model to study mechanisms that regulate inflammation. In particular, skin wound healing is dependent on a rapid, robust immune response and subsequent dampening of inflammatory signaling. While injury-induced inflammation has historically been attributed to keratinocytes and immune cells, a vast body of evidence supports the ability of non-immune cells to coordinate inflammation in numerous tissues and diseases. In this review, we concentrate on the active participation of tissue-resident adipocytes and fibroblasts in pro-inflammatory signaling after injury, and how altered cellular communication from these cells can contribute to irregular inflammation associated with aberrant wound healing. Furthering our understanding of how tissue-resident mesenchymal cells contribute to inflammation will likely reveal new targets that can be manipulated to regulate inflammation and repair.


Subject(s)
Adipocytes, White/immunology , Dermis/cytology , Dermis/injuries , Fibroblasts/immunology , Wound Healing/immunology , Aging/immunology , Aging/metabolism , Animals , Cell Communication/immunology , Cell Polarity/immunology , Cytokines/metabolism , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction/immunology
4.
Cell Stem Cell ; 28(3): 367-369, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33667356

ABSTRACT

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.


Subject(s)
COVID-19 , Career Mobility , Research Personnel , Stem Cells , Humans
5.
Cell Stem Cell ; 26(6): 880-895.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32302523

ABSTRACT

Mature adipocytes store fatty acids and are a common component of tissue stroma. Adipocyte function in regulating bone marrow, skin, muscle, and mammary gland biology is emerging, but the role of adipocyte-derived lipids in tissue homeostasis and repair is poorly understood. Here, we identify an essential role for adipocyte lipolysis in regulating inflammation and repair after injury in skin. Genetic mouse studies revealed that dermal adipocytes are necessary to initiate inflammation after injury and promote subsequent repair. We find through histological, ultrastructural, lipidomic, and genetic experiments in mice that adipocytes adjacent to skin injury initiate lipid release necessary for macrophage inflammation. Tamoxifen-inducible genetic lineage tracing of mature adipocytes and single-cell RNA sequencing revealed that dermal adipocytes alter their fate and generate ECM-producing myofibroblasts within wounds. Thus, adipocytes regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.


Subject(s)
Lipolysis , Myofibroblasts , Adipocytes , Animals , Macrophages , Mice , Skin
6.
Science ; 362(6417)2018 11 23.
Article in English | MEDLINE | ID: mdl-30467144

ABSTRACT

During tissue repair, myofibroblasts produce extracellular matrix (ECM) molecules for tissue resilience and strength. Altered ECM deposition can lead to tissue dysfunction and disease. Identification of distinct myofibroblast subsets is necessary to develop treatments for these disorders. We analyzed profibrotic cells during mouse skin wound healing, fibrosis, and aging and identified distinct subpopulations of myofibroblasts, including adipocyte precursors (APs). Multiple mouse models and transplantation assays demonstrate that proliferation of APs but not other myofibroblasts is activated by CD301b-expressing macrophages through insulin-like growth factor 1 and platelet-derived growth factor C. With age, wound bed APs and differential gene expression between myofibroblast subsets are reduced. Our findings identify multiple fibrotic cell populations and suggest that the environment dictates functional myofibroblast heterogeneity, which is driven by fibroblast-immune interactions after wounding.


Subject(s)
Macrophages/physiology , Myofibroblasts/physiology , Re-Epithelialization/physiology , Skin/injuries , Wound Healing/physiology , Adipocytes/physiology , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Fibrosis , Integrin beta1/genetics , Keloid/pathology , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Lymphokines/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Platelet-Derived Growth Factor/metabolism , Re-Epithelialization/genetics , Skin/immunology , Skin/pathology , Skin Aging/physiology , Transcriptome , Wound Healing/genetics
7.
Nat Commun ; 9(1): 3592, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30181538

ABSTRACT

Adipocytes undergo pronounced changes in size and behavior to support diverse tissue functions, but the mechanisms that control these changes are not well understood. Mammary gland-associated white adipose tissue (mgWAT) regresses in support of milk fat production during lactation and expands during the subsequent involution of milk-producing epithelial cells, providing one of the most marked physiological examples of adipose growth. We examined cellular mechanisms and functional implications of adipocyte and lipid dynamics in the mouse mammary gland (MG). Using in vivo analysis of adipocyte precursors and genetic tracing of mature adipocytes, we find mature adipocyte hypertrophy to be a primary mechanism of mgWAT expansion during involution. Lipid tracking and lipidomics demonstrate that adipocytes fill with epithelial-derived milk lipid. Furthermore, ablation of mgWAT during involution reveals an essential role for adipocytes in milk trafficking from, and proper restructuring of, the mammary epithelium. This work advances our understanding of MG remodeling and tissue-specific roles for adipocytes.


Subject(s)
Adipocytes/cytology , Lipid Metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Adipocytes/metabolism , Adipocytes, White/cytology , Adipocytes, White/physiology , Animals , Breast Feeding , Cell Size , Epithelial Cells/cytology , Epithelial Cells/physiology , Fatty Acids/metabolism , Female , Humans , Lactation/physiology , Mammary Glands, Animal/physiology , Mammary Glands, Human/physiology , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
9.
Cell Metab ; 25(3): 493-494, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28273471

ABSTRACT

Excess ECM and fibrosis of white adipose tissue (WAT) is associated with tissue dysfunction and type 2 diabetes. In this issue of Cell Metabolism, Marcelin et al. (2017) elucidate a key mechanism behind WAT fibrosis in which the activation of PDGFRα on adipocyte precursors drives this population toward a fibrotic phenotype.


Subject(s)
Adipose Tissue, White/cytology , Diabetes Mellitus, Type 2 , Adipocytes/cytology , Fibrosis , Humans , Phenotype
10.
Cell Stem Cell ; 19(6): 738-751, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27746098

ABSTRACT

Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.


Subject(s)
Adipocytes/cytology , Adipocytes/enzymology , Cell Self Renewal , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Skin/cytology , Stem Cells/cytology , Adipogenesis , Animals , CD24 Antigen/metabolism , Cell Proliferation , Dermis/metabolism , Gene Expression Profiling , Hyperplasia , Mice, Inbred C57BL , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
11.
J Invest Dermatol ; 136(9): 1885-1891, 2016 09.
Article in English | MEDLINE | ID: mdl-27287183

ABSTRACT

Regeneration of skin's barrier function after injury requires temporally coordinated cellular interactions between multiple cell types. Macrophages are essential inflammatory cells in skin wound regeneration. These cells switch their phenotype from inflammatory in the early regenerative stages to anti-inflammatory in the midstages of healing to coordinate skin repair. However, little is known about how different subsets of anti-inflammatory macrophages contribute to skin wound healing. Here, we characterize midstage macrophages (CD45(+)/CD11b(+)/F4-80(+)) and identify two major populations: CD206(+)/CD301b(+) and CD206(+)/CD301b(-). The numbers of CD206(+)/CD301b(+) macrophages increased concomitantly with repair, when the anti-inflammatory phenotype switch occurs in midstage healing. Using diphtheria toxin-mediated depletion models in mice, we show that selective depletion of midstage CD301b-expressing macrophages phenocopied wound healing defects observed in mice where multiple myeloid lineages are depleted. Additionally, when FACS-isolated subpopulations of myeloid cells were transplanted into 3-day wounds of syngeneic mice, only CD206(+)/CD301b(+) macrophages significantly increased proliferation and fibroblast repopulation. These data show that the CD301b-expressing subpopulation of macrophages is critical for activation of reparative processes during the midstage of cutaneous repair.


Subject(s)
Lectins, C-Type/metabolism , Skin/injuries , Skin/metabolism , Wound Healing/physiology , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Sensitivity and Specificity
12.
Annu Rev Cell Dev Biol ; 32: 609-631, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27146311

ABSTRACT

Classically, white adipose tissue (WAT) was considered an inert component of connective tissue but is now appreciated as a major regulator of metabolic physiology and endocrine homeostasis. Recent work defining how WAT develops and expands in vivo emphasizes the importance of specific locations of WAT or depots in metabolic regulation. Interestingly, mature white adipocytes are integrated into several tissues. A new perspective regarding the in vivo regulation and function of WAT in these tissues has highlighted an essential role of adipocytes in tissue homeostasis and regeneration. Finally, there has been significant progress in understanding how mature adipocytes regulate the pathology of several diseases. In this review, we discuss these novel roles of WAT in the homeostasis and regeneration of epithelial, muscle, and immune tissues and how they contribute to the pathology of several disorders.


Subject(s)
Adipocytes/metabolism , Organogenesis , Regeneration/physiology , Stem Cell Niche , Animals , Disease , Humans , Models, Biological
13.
J Vis Exp ; (99): e52328, 2015 May 19.
Article in English | MEDLINE | ID: mdl-26068121

ABSTRACT

The ventricular system carries and circulates cerebral spinal fluid (CSF) and facilitates clearance of solutes and toxins from the brain. The functional units of the ventricles are ciliated epithelial cells termed ependymal cells, which line the ventricles and through ciliary action are capable of generating laminar flow of CSF at the ventricle surface. This monolayer of ependymal cells also provides barrier and filtration functions that promote exchange between brain interstitial fluids (ISF) and circulating CSF. Biochemical changes in the brain are thereby reflected in the composition of the CSF and destruction of the ependyma can disrupt the delicate balance of CSF and ISF exchange. In humans there is a strong correlation between lateral ventricle expansion and aging. Age-associated ventriculomegaly can occur even in the absence of dementia or obstruction of CSF flow. The exact cause and progression of ventriculomegaly is often unknown; however, enlarged ventricles can show regional and, often, extensive loss of ependymal cell coverage with ventricle surface astrogliosis and associated periventricular edema replacing the functional ependymal cell monolayer. Using MRI scans together with postmortem human brain tissue, we describe how to prepare, image and compile 3D renderings of lateral ventricle volumes, calculate lateral ventricle volumes, and characterize periventricular tissue through immunohistochemical analysis of en face lateral ventricle wall tissue preparations. Corresponding analyses of mouse brain tissue are also presented supporting the use of mouse models as a means to evaluate changes to the lateral ventricles and periventricular tissue found in human aging and disease. Together, these protocols allow investigations into the cause and effect of ventriculomegaly and highlight techniques to study ventricular system health and its important barrier and filtration functions within the brain.


Subject(s)
Lateral Ventricles/anatomy & histology , Age Factors , Animals , Disease Models, Animal , Ependyma/anatomy & histology , Ependyma/cytology , Ependyma/pathology , Epithelial Cells/cytology , Epithelial Cells/pathology , Gliosis/pathology , Humans , Hydrocephalus/pathology , Imaging, Three-Dimensional/methods , Lateral Ventricles/cytology , Lateral Ventricles/pathology , Magnetic Resonance Imaging , Mice , Models, Anatomic , Neuroglia/cytology , Neuroglia/pathology
14.
Adipocyte ; 3(3): 206-11, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25068087

ABSTRACT

The study of adipose tissue in vivo has been significantly advanced through the use of genetic mouse models. While the aP2-Cre(BI) and aP2-Cre(Salk) lines have been widely used to target adipose tissue, the specificity of these lines for adipocytes has recently been questioned. Here we characterize Cre recombinase activity in multiple cell populations of the major adipose tissue depots of these and other Cre lines using the membrane-Tomato/membrane-GFP (mT/mG) dual fluorescent reporter. We find that the aP2-Cre(BI) and aP2-Cre(Salk) lines lack specificity for adipocytes within adipose tissue, and that the aP2-Cre(BI) line does not efficiently target adipocytes in white adipose depots. Alternatively, the Adiponectin-CreERT line shows high efficiency and specificity for adipocytes, while the PdgfRα-CreERUCL and PdgfRα-CreERJHU lines do not efficiently target adipocyte precursor cells in the major adipose depots. Instead, we show that the PdgfRα-Cre line is preferable for studies targeting adipocyte precursor cells in vivo.

15.
Article in English | MEDLINE | ID: mdl-24591537

ABSTRACT

Adipocytes are intimately associated with the dermal compartment of the skin, existing in a specialized dermal depot and displaying dynamic changes in size during tissue homeostasis. However, the roles of adipocytes in cutaneous biology and disease are not well understood. Traditionally, adipocytes within tissues were thought to act as reservoirs of energy, as thermal, or as structural support. In this review, we discuss recent studies revealing the cellular basis of the dynamic development and regenerative capacity of dermal adipocytes associated with the hair cycle and following injury. We discuss and speculate on potential roles of dermal adipocytes in cutaneous biology with an emphasis on communication during hair follicle growth and wound healing. Finally, we explore how alterations in the dermal adipose tissue may support clinical manifestations of cutaneous diseases such as lipodystrophy, obesity, and alopecia.


Subject(s)
Adipocytes/physiology , Skin Diseases/pathology , Skin/cytology , Adipocytes/cytology , Hair/physiology , Hair Follicle/cytology , Homeostasis/physiology , Hormones/physiology , Humans , Regeneration/physiology , Skin Aging/physiology , Wound Healing/physiology
16.
Aging Cell ; 13(2): 340-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24341850

ABSTRACT

Age-associated ventriculomegaly is typically attributed to neurodegeneration; however, additional factors might initiate or contribute to progressive ventricular expansion. By directly linking postmortem human MRI sequences with histological features of periventricular tissue, we show that substantial lateral ventricle surface gliosis is associated with ventriculomegaly. To examine whether loss of ependymal cell coverage resulting in ventricle surface glial scarring can lead directly to ventricle enlargement independent of any other injury or degenerative loss, we modeled in mice the glial scarring found along the lateral ventricle surface in aged humans. Neuraminidase, which cleaves glycosidic linkages of apical adherens junction proteins, was administered intracerebroventricularly to denude areas of ependymal cells. Substantial ependymal cell loss resulted in reactive gliosis rather than stem cell-mediated regenerative repair of the ventricle lining, and the gliotic regions showed morphologic and phenotypic characteristics similar to those found in aged humans. Increased levels of aquaporin-4, indicative of edema, observed in regions of periventricular gliosis in human tissue were also replicated in our mouse model. 3D modeling together with volume measurements revealed that mice with ventricle surface scarring developed expanded ventricles, independent of neurodegeneration. Through a comprehensive, comparative analysis of the lateral ventricles and associated periventricular tissue in aged humans and mouse, followed by modeling of surface gliosis in mice, we have demonstrated a direct link between lateral ventricle surface gliosis and ventricle enlargement. These studies highlight the importance of maintaining an intact ependymal cell lining throughout aging.


Subject(s)
Aging/pathology , Cerebral Ventricles/abnormalities , Ependyma/pathology , Gliosis/complications , Gliosis/pathology , Adult , Aged , Aged, 80 and over , Animals , Aquaporin 4/metabolism , Cerebral Ventricles/pathology , Disease Models, Animal , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Middle Aged , Neuraminidase/metabolism , Organ Size , Postmortem Changes , Stem Cells/pathology , Up-Regulation , Young Adult
17.
Nat Genet ; 45(9): 995-1003, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23872636

ABSTRACT

DYX1C1 has been associated with dyslexia and neuronal migration in the developing neocortex. Unexpectedly, we found that deleting exons 2-4 of Dyx1c1 in mice caused a phenotype resembling primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease, laterality defects and male infertility. This phenotype was confirmed independently in mice with a Dyx1c1 c.T2A start-codon mutation recovered from an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Morpholinos targeting dyx1c1 in zebrafish also caused laterality and ciliary motility defects. In humans, we identified recessive loss-of-function DYX1C1 mutations in 12 individuals with PCD. Ultrastructural and immunofluorescence analyses of DYX1C1-mutant motile cilia in mice and humans showed disruptions of outer and inner dynein arms (ODAs and IDAs, respectively). DYX1C1 localizes to the cytoplasm of respiratory epithelial cells, its interactome is enriched for molecular chaperones, and it interacts with the cytoplasmic ODA and IDA assembly factor DNAAF2 (KTU). Thus, we propose that DYX1C1 is a newly identified dynein axonemal assembly factor (DNAAF4).


Subject(s)
Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Cilia/genetics , Cilia/metabolism , Nerve Tissue Proteins/genetics , Animals , Cilia/ultrastructure , Disease Models, Animal , Ependyma/metabolism , Ependyma/pathology , Gene Knockdown Techniques , Gene Order , Gene Targeting , Humans , Intracellular Space/metabolism , Kartagener Syndrome/genetics , Kartagener Syndrome/metabolism , Male , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Protein Binding , Protein Transport , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Zebrafish
18.
J Neurosci ; 32(20): 6947-56, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22593063

ABSTRACT

Through adulthood, the rodent subventricular zone (SVZ) stem cell niche generates new olfactory bulb interneurons. We had previously reported that the number of new neurons produced in the SVZ declines through aging; however, age-related changes attributable specifically to the SVZ neural stem cell (NSC) population have not been fully characterized. Here, we conducted a spatiotemporal evaluation of adult SVZ NSCs. We assessed ventricle-contacting NSCs, which together with ependymal cells form regenerative units (pinwheels) along the lateral wall of the lateral ventricle. Based on their apical GFAP-expressing process, individual NSCs were identified across the ventricle surface using serial reconstruction of the SVZ. We observed an 86% decline in total NSCs/mm² of intact ependyma in 2-year old versus 3-month-old mice, with fewer NSC processes within each aged pinwheel. This resulted in an associated 78% decline in total pinwheel units/mm². Regional analysis along the lateral ventricle surface revealed that the age-dependent decline of NSCs and pinwheels is spatially uniform and ultimately maintains the conserved ratio of olfactory bulb interneuron subtypes generated in young mice. However, the overall neurogenic output of the aged SVZ is reduced. Surprisingly, we found no significant change in the number of actively proliferating NSCs per mm² of ventricle surface. Instead, our data reveal that, although the total NSC number, pinwheel units and NSCs per pinwheel decline with age, the percentage of actively, mitotic NSCs increases, indicating that age-related declines in SVZ-mediated olfactory bulb neurogenesis occur downstream of NSC proliferation.


Subject(s)
Aging/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Cell Proliferation , Constriction, Pathologic/physiopathology , Interneurons/physiology , Lateral Ventricles/cytology , Male , Mice , Mice, Inbred ICR , Neural Stem Cells/cytology , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Bulb/physiology
19.
Aging Dis ; 2(1): 149-163, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-22140636

ABSTRACT

The persistence of an active subventricular zone neural stem cell niche in the adult mammalian forebrain supports its continued role in the production of new neurons and in generating cells to function in repair through adulthood. Unfortunately, with increasing age the niche begins to deteriorate, compromising these functions. The reasons for this decline are not clear. Studies are beginning to define the molecular and physiologic changesin the microenvironment of the aging subventricular zone niche. New revelations from aging studies will allow for a more thorough understanding of what reparative functions are lost in the aged brain, the progression of niche demise and the possibility for therauptic intervention to improve aging brain function.

20.
Aging Dis ; 2(1): 49-63, 2011 Feb.
Article in English | MEDLINE | ID: mdl-22396866

ABSTRACT

The persistence of an active subventricular zone neural stem cell niche in the adult mammalian forebrain supports its continued role in the production of new neurons and in generating cells to function in repair through adulthood. Unfortunately, with increasing age the niche begins to deteriorate, compromising these functions. The reasons for this decline are not clear. Studies are beginning to define the molecular and physiologic changes in the microenvironment of the aging subventricular zone niche. New revelations from aging studies will allow for a more thorough understanding of which reparative functions are lost in the aged brain, the progression of niche demise and the possibility for therauptic intervention to improve aging brain function.

SELECTION OF CITATIONS
SEARCH DETAIL
...