Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Genes Dev ; 34(23-24): 1713-1734, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184221

ABSTRACT

Through recurrent bouts synchronous with the hair cycle, quiescent melanocyte stem cells (McSCs) become activated to generate proliferative progeny that differentiate into pigment-producing melanocytes. The signaling factors orchestrating these events remain incompletely understood. Here, we use single-cell RNA sequencing with comparative gene expression analysis to elucidate the transcriptional dynamics of McSCs through quiescence, activation, and melanocyte maturation. Unearthing converging signs of increased WNT and BMP signaling along this progression, we endeavored to understand how these pathways are integrated. Employing conditional lineage-specific genetic ablation studies in mice, we found that loss of BMP signaling in the lineage leads to hair graying due to a block in melanocyte maturation. We show that interestingly, BMP signaling functions downstream from activated McSCs and maintains WNT effector, transcription factor LEF1. Employing pseudotime analysis, genetics, and chromatin landscaping, we show that following WNT-mediated activation of McSCs, BMP and WNT pathways collaborate to trigger the commitment of proliferative progeny by fueling LEF1- and MITF-dependent differentiation. Our findings shed light upon the signaling interplay and timing of cues that orchestrate melanocyte lineage progression in the hair follicle and underscore a key role for BMP signaling in driving complete differentiation.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/genetics , Melanocytes/cytology , Signal Transduction/genetics , Stem Cells/cytology , Animals , Cell Lineage/genetics , Gene Expression Profiling , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Single-Cell Analysis
2.
Development ; 147(9)2020 05 05.
Article in English | MEDLINE | ID: mdl-32198156

ABSTRACT

Murine muscle stem cells (MuSCs) experience a transition from quiescence to activation that is required for regeneration, but it remains unknown if the trajectory and dynamics of activation change with age. Here, we use time-lapse imaging and single cell RNA-seq to measure activation trajectories and rates in young and aged MuSCs. We find that the activation trajectory is conserved in aged cells, and we develop effective machine-learning classifiers for cell age. Using cell-behavior analysis and RNA velocity, we find that activation kinetics are delayed in aged MuSCs, suggesting that changes in stem cell dynamics may contribute to impaired stem cell function with age. Intriguingly, we also find that stem cell activation appears to be a random walk-like process, with frequent reversals, rather than a continuous linear progression. These results support a view of the aged stem cell phenotype as a combination of differences in the location of stable cell states and differences in transition rates between them.


Subject(s)
Cellular Senescence/physiology , Muscle, Skeletal/metabolism , Stem Cells/metabolism , Animals , Cells, Cultured , Immunohistochemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , RNA-Seq , Stem Cells/cytology , Time-Lapse Imaging
3.
EMBO J ; 37(9)2018 05 02.
Article in English | MEDLINE | ID: mdl-29615452

ABSTRACT

Tissues contain distinct stem cell niches, but whether cell turnover is coordinated between niches during growth is unknown. Here, we report that in mouse skin, hair growth is accompanied by sebaceous gland and interfollicular epidermis expansion. During hair growth, cells in the bulge and outer root sheath temporarily upregulate the glutamate transporter SLC1A3, and the number of SLC1A3+ basal cells in interfollicular epidermis and sebaceous gland increases. Fate mapping of SLC1A3+ cells in mice revealed transient expression in proliferating stem/progenitor cells in all three niches. Deletion of slc1a3 delays hair follicle anagen entry, uncouples interfollicular epidermis and sebaceous gland expansion from the hair cycle, and leads to reduced fur density in aged mice, indicating a role of SLC1A3 in stem/progenitor cell activation. Modulation of metabotropic glutamate receptor 5 activity mimics the effects of SLC1A3 deletion or inhibition. These data reveal that stem/progenitor cell activation is synchronized over distinct niches during growth and identify SLC1A3 as a general marker and effector of activated epithelial stem/progenitor cells throughout the skin.


Subject(s)
Cell Proliferation/physiology , Epidermis/growth & development , Excitatory Amino Acid Transporter 1/biosynthesis , Gene Expression Regulation/physiology , Sebaceous Glands/growth & development , Stem Cells/metabolism , Animals , Excitatory Amino Acid Transporter 1/genetics , Mice , Mice, Transgenic , Sebaceous Glands/cytology
4.
Neurourol Urodyn ; 38(8): 2140-2150, 2019 11.
Article in English | MEDLINE | ID: mdl-31452249

ABSTRACT

AIM: To investigate the possibility and mechanism of microenergy acoustic pulses (MAP) for activating tissue resident stem/progenitor cells within pelvic and urethral muscle and possible mechanism. METHODS: The female Zucker Lean and Zucker Fatty rats were randomly divided into four groups: ZL control, ZLMAP, ZF control, and ZFMAP. MAP was applied at 0.033 mJ/mm2 , 3 Hz for 500 pulses, and the urethra and pelvic floor muscles of each rat was then harvested for cell isolation and flow cytometry assay. Freshly isolated cells were analyzed by flow cytometry for Pax-7, Int-7α, H3P, and EdU expression. Meanwhile, pelvic floor muscle-derived stem cells (MDSCs) were harvested through magnetic-activated cell sorting, MAP was then applied to MDSCs to assess the mechanism of stem cell activation. RESULTS: Obesity reduced EdU-label-retaining cells and satellite cells in both pelvic floor muscle and urethra, while MAP activated those cells and enhanced cell proliferation, which promoted regeneration of striated muscle cells of the pelvic floor and urethral sphincter. Activation of focal adhesion kinase (FAK)/AMP-activated protein kinase (AMPK) /Wnt/ß-catenin signaling pathways by MAP is the potential mechanism. CONCLUSIONS: MAP treatment activated tissue resident stem cells within pelvic floor and urethral muscle in situ via activating FAK-AMPK and Wnt/ß-catenin signaling pathway.


Subject(s)
Muscle, Skeletal/physiology , Obesity/physiopathology , Pelvic Floor/physiopathology , Satellite Cells, Skeletal Muscle/physiology , Urethra/physiopathology , Urinary Incontinence, Stress/physiopathology , Acoustic Stimulation , Acoustics , Animals , Antigens, CD/metabolism , Cell Proliferation , Deoxyuridine , Disease Models, Animal , Female , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alpha Chains/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/cytology , Muscle, Striated/cytology , Muscle, Striated/physiology , Myoblasts/physiology , Obesity/complications , Paired Box Transcription Factors , Rats , Rats, Zucker , Regeneration , Stem Cells , Urethra/cytology , Urinary Incontinence, Stress/etiology , Wnt Signaling Pathway
5.
Mater Today Bio ; 20: 100664, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37251417

ABSTRACT

Bone defects in patients entail the microenvironment that needs to boost the functions of stem cells (e.g., proliferation, migration, and differentiation) while alleviating severe inflammation induced by high oxidative stress. Biomaterials can help to shift the microenvironment by regulating these multiple events. Here we report multifunctional composite hydrogels composed of photo-responsive Gelatin Methacryloyl (GelMA) and dendrimer (G3)-functionalized nanoceria (G3@nCe). Incorporation of G3@nCe into GelMA could enhance the mechanical properties of hydrogels and their enzymatic ability to clear reactive oxygen species (ROS). The G3@nCe/GelMA hydrogels supported the focal adhesion of mesenchymal stem cells (MSCs) and further increased their proliferation and migration ability (vs. pristine GelMA and nCe/GelMA). Moreover, the osteogenic differentiation of MSCs was significantly stimulated upon the G3@nCe/GelMA hydrogels. Importantly, the capacity of G3@nCe/GelMA hydrogels to scavenge extracellular ROS enabled MSCs to survive against H2O2-induced high oxidative stress. Transcriptome analysis by RNA sequencing identified the genes upregulated and the signalling pathways activated by G3@nCe/GelMA that are associated with cell growth, migration, osteogenesis, and ROS-metabolic process. When implanted subcutaneously, the hydrogels exhibited excellent tissue integration with a sign of material degradation while the inflammatory response was minimal. Furthermore, G3@nCe/GelMA hydrogels demonstrated effective bone regeneration capacity in a rat critical-sized bone defect model, possibly due to an orchestrated capacity of enhancing cell proliferation, motility and osteogenesis while alleviating oxidative stress.

6.
Heliyon ; 9(9): e20182, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37809421

ABSTRACT

Chromosomal instability (CIN) is critical for tumor evolution, yet its relationship with stemness is unclear. Here, we describe CIN as a key stress induced during tumor initiation that is uniquely tolerated by breast cancer stem cells in an activated signaling state (aCSCs). While we noted elevated CIN specifically in tumors from aCSCs, this was not intrinsic to these cells, as baseline levels were similar to non-stem cell types. This suggests that CIN is induced during tumor initiation, and that aCSCs can better tolerate this stress. Further, this increased CIN may be transient, as it was only in low-burden aCSC tumors, with levels diminishing in more established disease. Phospho-array profiling revealed specific activation of c-Jun stress signaling in aCSCs, which we hypothesized could induce genes responsible for CIN tolerance. Indeed, we identified AXL as a c-Jun dependent gene enriched in aCSCs that enhances resistance to this stress. Thus, CIN tolerance mediated by c-Jun/AXL signaling may be a defining feature of stemness, contributing to breast cancer progression.

7.
Chin Med ; 18(1): 84, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37454125

ABSTRACT

BACKGROUND: As a supplement for promoting hair health, Shi-Bi-Man (SBM) is a prescription comprising various traditional Chinese medicines. Though SBM has been reported to promote hair regeneration, its molecular mechanism remains unclear. Cynomolgus monkeys (Macaca fascicularis) are non-human primates with a gene expression profile similar to that of humans. The purpose of this research is to evaluate the effect of SBM on promoting hair regeneration in cynomolgus monkeys and to reveal the underlying mechanism. METHODS: The effect of SBM on hair regeneration was observed by skin administration on 6 cynomolgus monkeys with artificial back shaving. The molecular mechanism of SBM was studied using single-cell RNA sequencing (scRNA-seq) in combination with quantitative polymerase chain reaction (qPCR) detection for gene transcription level, and immunofluorescence staining verification for protein level. RESULTS: SBM significantly induced hair regeneration in cynomolgus monkeys, increased hair follicle number and facilitated hair follicle development. ScRNA-seq revealed an increase in the number of hair follicle stem cells (HFSCs) with a higher activation state, as evidenced by the higher expression of activation marker LDHA related to metabolism and the proliferation marker MKI67. Immunofluorescence analysis at the protein level and qPCR at the mRNA level confirmed the sequencing data. Cellchat analysis revealed an enrichment of ligand-receptor pairs involved in intercellular communication in Laminin-related pathways. CONCLUSION: SBM significantly promotes hair regeneration in cynomolgus monkeys. Mechanically, SBM can up-regulate LDHA-mediated lactic acid metabolism and drive HFSC activation, which in turn promotes the proliferation and differentiation of HFSCs.

8.
Stem Cell Reports ; 18(5): 1227-1243, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37084727

ABSTRACT

The molecular mechanisms allowing hair follicles to periodically activate their stem cells (HFSCs) are incompletely characterized. Here, we identify the transcription factor IRX5 as a promoter of HFSC activation. Irx5-/- mice have delayed anagen onset, with increased DNA damage and diminished HFSC proliferation. Open chromatin regions form near cell cycle progression and DNA damage repair genes in Irx5-/- HFSCs. DNA damage repair factor BRCA1 is an IRX5 downstream target. Inhibition of FGF kinase signaling partially rescues the anagen delay in Irx5-/- mice, suggesting that the Irx5-/- HFSC quiescent phenotype is partly due to failure to suppress Fgf18 expression. Interfollicular epidermal stem cells also show decreased proliferation and increased DNA damage in Irx5-/-mice. Consistent with a role for IRX5 as a promoter of DNA damage repair, we find that IRX genes are upregulated in many cancer types and that there is a correlation between IRX5 and BRCA1 expression in breast cancer.


Subject(s)
Hair Follicle , Stem Cells , Mice , Animals , Hair Follicle/metabolism , Stem Cells/metabolism , Signal Transduction , Gene Expression Regulation , DNA Damage , Transcription Factors/genetics , Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
9.
Cell Rep ; 41(1): 111447, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36198277

ABSTRACT

Respiratory tract infections are among the deadliest communicable diseases worldwide. Severe cases of viral lung infections are often associated with a cytokine storm and alternating platelet numbers. We report that hematopoietic stem and progenitor cells (HSPCs) sense a non-systemic influenza A virus (IAV) infection via inflammatory cytokines. Irrespective of antiviral treatment or vaccination, at a certain threshold of IAV titer in the lung, CD41-positive hematopoietic stem cells (HSCs) enter the cell cycle while endothelial protein C receptor-positive CD41-negative HSCs remain quiescent. Active CD41-positive HSCs represent the source of megakaryocytes, while their multi-lineage reconstitution potential is reduced. This emergency megakaryopoiesis is thrombopoietin independent and attenuated in IAV-infected interleukin-1 receptor-deficient mice. Newly produced platelets during IAV infection are immature and hyper-reactive. After viral clearance, HSC quiescence is re-established. Our study reveals that non-systemic viral respiratory infection has an acute impact on HSCs via inflammatory cytokines to counteract IAV-induced thrombocytopenia.


Subject(s)
Influenza A virus , Influenza, Human , Animals , Antiviral Agents/metabolism , Cytokines/metabolism , Endothelial Protein C Receptor/metabolism , Hematopoiesis , Humans , Influenza, Human/metabolism , Megakaryocytes/metabolism , Mice , Receptors, Interleukin-1/metabolism , Thrombopoietin/metabolism
10.
Cell Stem Cell ; 29(6): 933-947.e6, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35597234

ABSTRACT

Many tissues harbor quiescent stem cells that are activated upon injury, subsequently proliferating and differentiating to repair tissue damage. Mechanisms by which stem cells sense injury and transition from quiescence to activation, however, remain largely unknown. Resident skeletal muscle stem cells (MuSCs) are essential orchestrators of muscle regeneration and repair. Here, with a combination of in vivo and ex vivo approaches, we show that quiescent MuSCs have elaborate, Rac GTPase-promoted cytoplasmic projections that respond to injury via the upregulation of Rho/ROCK signaling, facilitating projection retraction and driving downstream activation events. These early events involve rapid cytoskeletal rearrangements and occur independently of exogenous growth factors. This mechanism is conserved across a broad range of MuSC activation models, including injury, disease, and genetic loss of quiescence. Our results redefine MuSC activation and present a central mechanism by which quiescent stem cells initiate responses to injury.


Subject(s)
Satellite Cells, Skeletal Muscle , rho GTP-Binding Proteins , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal , Myoblasts/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/metabolism , rho GTP-Binding Proteins/metabolism
11.
Cell Stem Cell ; 29(9): 1315-1332.e9, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35998642

ABSTRACT

Quiescence regulation is essential for adult stem cell maintenance and sustained regeneration. Our studies uncovered that physiological changes in mitochondrial shape regulate the quiescent state of adult muscle stem cells (MuSCs). We show that MuSC mitochondria rapidly fragment upon an activation stimulus, via systemic HGF/mTOR, to drive the exit from deep quiescence. Deletion of the mitochondrial fusion protein OPA1 and mitochondrial fragmentation transitions MuSCs into G-alert quiescence, causing premature activation and depletion upon a stimulus. OPA1 loss activates a glutathione (GSH)-redox signaling pathway promoting cell-cycle progression, myogenic gene expression, and commitment. MuSCs with chronic OPA1 loss, leading to mitochondrial dysfunction, continue to reside in G-alert but acquire severe cell-cycle defects. Additionally, we provide evidence that OPA1 decline and impaired mitochondrial dynamics contribute to age-related MuSC dysfunction. These findings reveal a fundamental role for OPA1 and mitochondrial dynamics in establishing the quiescent state and activation potential of adult stem cells.


Subject(s)
Adult Stem Cells , Mitochondrial Proteins , Mitochondrial Dynamics , Muscles , Myoblasts
12.
Aging Cell ; 20(11): e13499, 2021 11.
Article in English | MEDLINE | ID: mdl-34687484

ABSTRACT

Neural stem cells (NSCs) in the adult and aged brain are largely quiescent, and require transcriptional reprogramming to re-enter the cell cycle. However, the mechanisms underlying these changes and how they are altered with age remain undefined. Here, we identify the chromatin accessibility differences between primary neural stem/progenitor cells in quiescent and activated states. These distinct cellular states exhibit shared and unique chromatin profiles, both associated with gene regulation. Accessible chromatin states specific to activation or quiescence are active enhancers bound by key pro-neurogenic and quiescence factors. In contrast, shared sites are enriched for core promoter elements associated with translation and metabolism. Unexpectedly, through integrated analysis, we find that many sites that become accessible during NSC activation are linked to gene repression and associated with pro-quiescence factors, revealing a novel mechanism that may preserve quiescence re-entry. Furthermore, we report that in aged NSCs, chromatin regions associated with metabolic and transcriptional functions bound by key pro-quiescence transcription factors lose accessibility, suggesting a novel mechanism of age-associated NSC dysfunction. Together, our findings reveal how accessible chromatin states regulate the transcriptional switch between NSC quiescence and activation, and how this switch is affected with age.


Subject(s)
Aging/genetics , Aging/metabolism , Cellular Senescence/genetics , Chromatin/genetics , Chromatin/metabolism , Neural Stem Cells/metabolism , Transcriptional Activation , Animals , Brain/cytology , Brain/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , Gene Expression Regulation , Gene Regulatory Networks , Histones/genetics , Histones/metabolism , Mice , Neurogenesis/genetics , Promoter Regions, Genetic/genetics , RNA-Seq/methods
13.
Cell Rep ; 32(6): 108022, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32783944

ABSTRACT

The transitions from developing to adult quiescent and activated neural stem cells (NSCs) are not well understood. Here, we use single-cell transcriptional profiling and lineage tracing to characterize these transitions in the murine forebrain. We show that the two forebrain NSC parental populations, embryonic cortex and ganglionic eminence radial precursors (RPs), are highly similar even though they make glutamatergic versus gabaergic neurons. Both RP populations progress linearly to transition from a highly active embryonic to a dormant adult stem cell state that still shares many similarities with embryonic RPs. When adult NSCs of either embryonic origin become reactivated to make gabaergic neurons, they acquire a developing ganglionic eminence RP-like identity. Thus, transitions from embryonic RPs to adult NSCs and back to neuronal progenitors do not involve fundamental changes in cell identity, but rather reflect conversions between activated and dormant NSC states that may be determined by the niche environment.


Subject(s)
Neural Stem Cells/metabolism , Neurogenesis/genetics , Prosencephalon/physiopathology , Animals , Cell Differentiation , Mice
14.
Am J Sports Med ; 48(12): 3112-3120, 2020 10.
Article in English | MEDLINE | ID: mdl-32045265

ABSTRACT

BACKGROUND: The competitive environment of athletics has promoted the exploration of any technology application that may offer an edge with performance and recovery from injury. Ischemic therapy is one such technology that has rapidly been incorporated into training rooms and physical therapy clinics worldwide. This therapy modality is reported to increase an athlete's ability to improve muscle mass, strength, and endurance. PURPOSE: To provide the sports medicine physician with an understanding of the current state of ischemic therapy technology, including treatment specifications, known physiological effects, hypothesized mechanisms, biochemical effects, athletic applications, medical applications, animal models, and future research recommendations. STUDY DESIGN: Literature review. METHODS: A computer-based search of the PubMed database was used to perform a comprehensive literature review on musculoskeletal ischemic therapy. RESULTS: The current research on ischemic therapy is largely composed of case series with varying equipment, methods, and therapy specifications. The publication of case series has value in identifying this technology for future research, but the results of these studies should not be justification for application to athletes without validation of safety and effectiveness. CONCLUSION: To date, ischemic therapy remains unvalidated, and the mechanism by which it improves muscle performance is not clear.


Subject(s)
Ischemic Preconditioning , Muscle, Skeletal/blood supply , Sports Medicine , Animals , Constriction , Humans
15.
Cell Stem Cell ; 25(3): 357-372.e7, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31230859

ABSTRACT

Inflammation coordinates tissue regeneration via damaged cell removal and stem cell activation. Hematopoietic stem cells (HSCs) survive inflammatory stress that kills other blood cells, but the mechanisms underlying this effect remain poorly understood. Here, we find that tumor necrosis factor α (TNF-α) acts differently on HSCs and progenitors, thus facilitating hematopoietic clearance and promoting regeneration. We show that while inducing myeloid progenitor apoptosis, TNF-α promotes HSC survival and myeloid differentiation by activating a strong and specific p65-nuclear factor κB (NF-κB)-dependent gene program that primarily prevents necroptosis rather than apoptosis, induces immunomodulatory functions, and poises HSCs for myeloid cell production. These TNF-α-driven mechanisms are critical for HSC response to inflammatory stress but are also hijacked in aged and malignant HSCs. Our results reveal several TNF-α-mediated pro-survival mechanisms unique to HSCs, highlight an important role for necroptosis in HSC killing, and establish TNF-α as a major pro-survival and pro-regeneration factor for HSCs.


Subject(s)
Hematopoietic Stem Cells/physiology , Inflammation/immunology , Myeloid Cells/physiology , Regeneration/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Self Renewal , Cell Survival , Gene Expression Regulation , Humans , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Necroptosis
16.
Cell Transplant ; 27(3): 485-500, 2018 03.
Article in English | MEDLINE | ID: mdl-29774769

ABSTRACT

Stroke induces complex and dynamic, local and systemic changes including inflammatory reactions, immune responses, and repair and recovery processes. Mesenchymal stem cells (MSCs) have been shown to enhance neurological recovery after stroke. We hypothesized that serum factors play a critical role in the activation of bone marrow (BM) MSCs after stroke such as by increasing proliferation, paracrine effects, and rejuvenation. Human MSCs (hMSCs) were grown in fetal bovine serum (FBS), normal healthy control serum (NS), or stroke patient serum (SS). MSCs cultured in growth medium with 10% SS or NS exhibited higher proliferation indices than those cultured with FBS ( P < 0.01). FBS-, NS-, and SS-hMSCs showed differences in the expression of trophic factors; vascular endothelial growth factor, glial cell-derived neurotrophic factor, and fibroblast growth factor were densely expressed in samples cultured with SS ( P < 0.01). In addition, SS-MSCs revealed different cell cycle- or aging-associated messenger RNA expression in a later passage, and ß-galactosidase staining showed the senescence of MSCs observed during culture expansion was lower in MSCs cultured with SS than those cultured with NS or FBS ( P < 0.01). Several proteins related to the activity of receptors, growth factors, and cytokines were more prevalent in the serum of stroke patients than in that of normal subjects. Neurogenesis and angiogenesis were markedly increased in rats that had received SS-MSCs ( P < 0.05), and these rats showed significant behavioral improvements ( P < 0.01). Our results indicate that stroke induces a process of recovery via the activation of MSCs. Culture methods for MSCs using SS obtained during the acute phase of a stroke could constitute a novel MSC activation method that is feasible and efficient for the neurorestoration of stroke.


Subject(s)
Bone Marrow Cells/cytology , Brain Ischemia/therapy , Ischemic Preconditioning/methods , Mesenchymal Stem Cells/cytology , Serum/metabolism , Stroke/therapy , Aged , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Infarction, Middle Cerebral Artery/therapy , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Reverse Transcriptase Polymerase Chain Reaction
17.
Cell Stem Cell ; 22(6): 879-892.e6, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29804890

ABSTRACT

Hematopoietic stem cells (HSCs) sustain hematopoiesis throughout life. HSCs exit dormancy to restore hemostasis in response to stressful events, such as acute blood loss, and must return to a quiescent state to prevent their exhaustion and resulting bone marrow failure. HSC activation is driven in part through the phosphatidylinositol 3-kinase (PI3K)/AKT/mTORC1 signaling pathway, but less is known about the cell-intrinsic pathways that control HSC dormancy. Here, we delineate an ERK-dependent, rate-limiting feedback mechanism that controls HSC fitness and their re-entry into quiescence. We show that the MEK/ERK and PI3K pathways are synchronously activated in HSCs during emergency hematopoiesis and that feedback phosphorylation of MEK1 by activated ERK counterbalances AKT/mTORC1 activation. Genetic or chemical ablation of this feedback loop tilts the balance between HSC dormancy and activation, increasing differentiated cell output and accelerating HSC exhaustion. These results suggest that MEK inhibitors developed for cancer therapy may find additional utility in controlling HSC activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Animals , Cells, Cultured , Coculture Techniques , Female , Humans , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Reactive Oxygen Species/metabolism
18.
Physiol Rep ; 5(11)2017 Jun.
Article in English | MEDLINE | ID: mdl-28596299

ABSTRACT

Aged skeletal muscle has an attenuated and delayed ability to proliferate satellite cells in response to resistance exercise. The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is a focal point for cell growth, however, the effect of postexercise mTORC1 activation on human skeletal muscle satellite cell (SC) proliferation is unknown. To test the proliferative capacity of skeletal muscle SC in aging muscle to a potent mTORC1 activator (i.e., EAA; essential amino acids) we recruited older (~72y) men to conduct leg resistance exercise (8setsx10reps) without (-EAA; n = 8) and with (+EAA: n = 11) ingestion of 10 g of EAA 1 h postexercise. Muscle biopsies were taken before exercise (Pre) and 24 h postexercise (Post) for assessment of expression and fiber type-specific Pax7+ SC, Ki67+Pax7+ SC and MyoD+ SC -EAA did not show an increase in Pax7+ satellite cells at Post(P > 0.82). Although statistical significance for an increase in Pax7 +  SC at 24 h post-RE was not observed in +EAA versus -EAA, we observed trends for a treatment difference (P < 0.1). When examining the change from Pre to Post trends were demonstrated (#/myofiber: P = 0.076; and %/myonuclei: P = 0.065) for a greater increase in +EAA versus -EAA Notably, we found an increase SC proliferation in +EAA, but not -EAA with increase in Ki67+ SC and MyoD+ cells (P < 0.05). Ki67+ SC also exhibited a significant group difference Post (P < 0.010). Pax7+ SC in fast twitch myofibers did not change and were not different between groups (P > 0.10). CDK2, MEF2C, RB1 mRNA only increased in +EAA (P < 0.05). Acute muscle satellite cell proliferative capacity may be partially rescued with postexercise EAA ingestion in older men.


Subject(s)
Amino Acids, Essential/pharmacology , Cell Proliferation , Muscle, Skeletal/drug effects , Resistance Training , Satellite Cells, Skeletal Muscle/drug effects , Aged , Amino Acids, Essential/administration & dosage , Case-Control Studies , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Dietary Supplements , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , MyoD Protein/genetics , MyoD Protein/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
19.
Cell Rep ; 19(3): 479-486, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423312

ABSTRACT

The activation of quiescent stem cells into the cell cycle is a key step in initiating the process of tissue repair. We recently reported that quiescent stem cells can transition into GAlert, a cellular state in which they have an increased functional ability to activate and participate in tissue repair. However, the precise molecular signals that induce GAlert in stem cells have remained elusive. Here, we show that the injury-induced regulation of hepatocyte growth factor (HGF) proteolytic processing via the systemic protease, hepatocyte growth factor activator (HGFA), stimulates GAlert in skeletal muscle stem cells (MuSCs) and fibro-adipogenic progenitors (FAPs). We demonstrate that administering active HGFA to animals is sufficient to induce GAlert in stem cells throughout the body and to significantly accelerate the processes of stem cell activation and tissue repair. Our data suggest that factors that induce GAlert will have broad therapeutic applications for regenerative medicine and wound healing.


Subject(s)
Cell Cycle/drug effects , Serine Endopeptidases/pharmacology , Stem Cells/cytology , Wound Healing/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , Animals , Fibroblasts/cytology , Fibroblasts/drug effects , Kinetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Serine Endopeptidases/administration & dosage , Serum/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
20.
Cell Rep ; 19(11): 2345-2356, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28614719

ABSTRACT

Quiescent long-term hematopoietic stem cells (LT-HSCs) are efficiently activated by type I interferon (IFN-I). However, this effect remains poorly investigated in the context of IFN-I-inducing virus infections. Here we report that both vesicular stomatitis virus (VSV) and murine cytomegalovirus (MCMV) infection induce LT-HSC activation that substantially differs from the effects triggered upon injection of synthetic IFN-I-inducing agents. In both infections, inflammatory responses had to exceed local thresholds within the bone marrow to confer LT-HSC cell cycle entry, and IFN-I receptor triggering was not critical for this activation. After resolution of acute MCMV infection, LT-HSCs returned to phenotypic quiescence. However, non-acute MCMV infection induced a sustained inflammatory milieu within the bone marrow that was associated with long-lasting impairment of LT-HSC function. In conclusion, our results show that systemic virus infections fundamentally affect LT-HSCs and that also non-acute inflammatory stimuli in bone marrow donors can affect the reconstitution potential of bone marrow transplants.


Subject(s)
Hematopoietic Stem Cells/metabolism , Infections/virology , Animals , Cell Cycle , Cell Proliferation , Hematopoietic Stem Cells/cytology , Mice , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL