Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.473
1.
Science ; 383(6687): eadi7342, 2024 Mar 08.
Article En | MEDLINE | ID: mdl-38452090

Lineage plasticity-a state of dual fate expression-is required to release stem cells from their niche constraints and redirect them to tissue compartments where they are most needed. In this work, we found that without resolving lineage plasticity, skin stem cells cannot effectively generate each lineage in vitro nor regrow hair and repair wounded epidermis in vivo. A small-molecule screen unearthed retinoic acid as a critical regulator. Combining high-throughput approaches, cell culture, and in vivo mouse genetics, we dissected its roles in tissue regeneration. We found that retinoic acid is made locally in hair follicle stem cell niches, where its levels determine identity and usage. Our findings have therapeutic implications for hair growth as well as chronic wounds and cancers, where lineage plasticity is unresolved.


Adult Stem Cells , Cell Plasticity , Epidermis , Hair Follicle , Tretinoin , Wound Healing , Animals , Mice , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Lineage/drug effects , Cell Lineage/physiology , Cell Plasticity/drug effects , Cell Plasticity/physiology , Epidermis/drug effects , Epidermis/physiology , Hair Follicle/cytology , Hair Follicle/drug effects , Hair Follicle/physiology , Tretinoin/metabolism , Tretinoin/pharmacology , Wound Healing/drug effects , Wound Healing/physiology , Rejuvenation/physiology , Cell Culture Techniques , Neoplasms/pathology , Mice, Inbred C57BL
2.
Stem Cell Reports ; 17(6): 1411-1427, 2022 06 14.
Article En | MEDLINE | ID: mdl-35523180

The insulin receptor (INSR) is an evolutionarily conserved signaling protein that regulates development and cellular metabolism. INSR signaling promotes neurogenesis in Drosophila; however, a specific role for the INSR in maintaining adult neural stem cells (NSCs) in mammals has not been investigated. We show that conditionally deleting the Insr gene in adult mouse NSCs reduces subventricular zone NSCs by ∼70% accompanied by a corresponding increase in progenitors. Insr deletion also produced hyposmia caused by aberrant olfactory bulb neurogenesis. Interestingly, hippocampal neurogenesis and hippocampal-dependent behaviors were unperturbed. Highly aggressive proneural and mesenchymal glioblastomas had high INSR/insulin-like growth factor (IGF) pathway gene expression, and isolated glioma stem cells had an aberrantly high ratio of INSR:IGF type 1 receptor. Moreover, INSR knockdown inhibited GBM tumorsphere growth. Altogether, these data demonstrate that the INSR is essential for a subset of normal NSCs, as well as for brain tumor stem cell self-renewal.


Adult Stem Cells , Lateral Ventricles/metabolism , Neural Stem Cells , Receptor, Insulin/metabolism , Somatomedins , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Lateral Ventricles/cytology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Somatomedins/metabolism
3.
Cell Rep ; 38(7): 110370, 2022 02 15.
Article En | MEDLINE | ID: mdl-35172139

The transition between quiescence and activation in neural stem and progenitor cells (NSPCs) is coupled with reversible changes in energy metabolism with key implications for lifelong NSPC self-renewal and neurogenesis. How this metabolic plasticity is ensured between NSPC activity states is unclear. We find that a state-specific rewiring of the mitochondrial proteome by the i-AAA peptidase YME1L is required to preserve NSPC self-renewal. YME1L controls the abundance of numerous mitochondrial substrates in quiescent NSPCs, and its deletion activates a differentiation program characterized by broad metabolic changes causing the irreversible shift away from a fatty-acid-oxidation-dependent state. Conditional Yme1l deletion in adult NSPCs in vivo results in defective self-renewal and premature differentiation, ultimately leading to NSPC pool depletion. Our results disclose an important role for YME1L in coordinating the switch between metabolic states of NSPCs and suggest that NSPC fate is regulated by compartmentalized changes in protein network dynamics.


Adult Stem Cells/metabolism , Cell Self Renewal , Metalloendopeptidases/metabolism , Mitochondria/enzymology , Neural Stem Cells/metabolism , Adult Stem Cells/cytology , Animals , Cell Proliferation , Citric Acid Cycle , Fatty Acids/metabolism , Gene Deletion , Metalloendopeptidases/deficiency , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Neural Stem Cells/cytology , Nucleotides/metabolism , Oxidation-Reduction , Proteolysis , Proteome/metabolism
4.
Cells ; 11(2)2022 01 10.
Article En | MEDLINE | ID: mdl-35053334

The ventricular-subventricular zone (V-SVZ) is a postnatal germinal niche. It holds a large population of neural stem cells (NSCs) that generate neurons and oligodendrocytes for the olfactory bulb and (primarily) the corpus callosum, respectively. These NSCs are heterogeneous and generate different types of neurons depending on their location. Positional identity among NSCs is thought to be controlled in part by intrinsic pathways. However, extrinsic cell signaling through the secreted ligand Sonic hedgehog (Shh) is essential for neurogenesis in both the dorsal and ventral V-SVZ. Here we used a genetic approach to investigate the role of the transcription factors GLI2 and GLI3 in the proliferation and cell fate of dorsal and ventral V-SVZ NSCs. We find that while GLI3 is expressed in stem cell cultures from both dorsal and ventral V-SVZ, the repressor form of GLI3 is more abundant in dorsal V-SVZ. Despite this high dorsal expression and the requirement for other Shh pathway members, GLI3 loss affects the generation of ventrally-, but not dorsally-derived olfactory interneurons in vivo and does not affect trilineage differentiation in vitro. However, loss of GLI3 in the adult dorsal V-SVZ in vivo results in decreased numbers of OLIG2-expressing progeny, indicating a role in gliogenesis.


Adult Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Zinc Finger Protein Gli3/metabolism , Adult Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Interneurons/metabolism , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/cytology , Smoothened Receptor/metabolism
5.
Int J Mol Sci ; 23(2)2022 Jan 07.
Article En | MEDLINE | ID: mdl-35054833

Currently, many different techniques exist for the surgical repair of peripheral nerves. The degree of injury dictates the repair and, depending on the defect or injury of the peripheral nerve, plastic surgeons can perform nerve repairs, grafts, and transfers. All the previously listed techniques are routinely performed in human patients, but a novel addition to these peripheral nerve surgeries involves concomitant fat grafting to the repair site at the time of surgery. Fat grafting provides adipose-derived stem cells to the injury site. Though fat grafting is performed as an adjunct to some peripheral nerve surgeries, there is no clear evidence as to which procedures have improved outcomes resultant from concomitant fat grafting. This review explores the evidence presented in various animal studies regarding outcomes of fat grafting at the time of various types of peripheral nerve surgery.


Adipose Tissue/cytology , Peripheral Nerve Injuries/surgery , Stem Cell Transplantation/methods , Adipose Tissue/transplantation , Adult Stem Cells/cytology , Animals , Disease Models, Animal , Humans , Nerve Regeneration , Peripheral Nerve Injuries/therapy
6.
Stem Cell Reports ; 17(1): 82-95, 2022 01 11.
Article En | MEDLINE | ID: mdl-35021050

Adult skeletal muscle stem cells (MuSCs) are important for muscle regeneration and constitute a potential source of cell therapy. However, upon isolation, MuSCs rapidly exit quiescence and lose transplantation potency. Maintenance of the quiescent state in vitro preserves MuSC transplantation efficiency and provides an opportunity to study the biology of quiescence. Here we show that Tubastatin A (TubA), an Hdac6 inhibitor, prevents primary cilium resorption, maintains quiescence, and enhances MuSC survival ex vivo. Phenotypic characterization and transcriptomic analysis of TubA-treated cells revealed that TubA maintains most of the biological features and molecular signatures of quiescence. Furthermore, TubA-treated MuSCs showed improved engraftment ability upon transplantation. TubA also induced a return to quiescence and improved engraftment of cycling MuSCs, revealing a potentially expanded application for MuSC therapeutics. Altogether, these studies demonstrate the ability of TubA to maintain MuSC quiescence ex vivo and to enhance the therapeutic potential of MuSCs and their progeny.


Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Cell Self Renewal/drug effects , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Muscle, Skeletal/cytology , Resting Phase, Cell Cycle/drug effects , Adult Stem Cells/metabolism , Animals , Cell Cycle , Cell Differentiation/drug effects , Gene Expression Profiling , Mice , Mice, Transgenic , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Stem Cell Transplantation , Transcriptome
7.
PLoS Genet ; 17(12): e1009609, 2021 12.
Article En | MEDLINE | ID: mdl-34898607

How and when potential becomes restricted in differentiating stem cell daughters is poorly understood. While it is thought that signals from the niche are actively required to prevent differentiation, another model proposes that stem cells can reversibly transit between multiple states, some of which are primed, but not committed, to differentiate. In the Drosophila testis, somatic cyst stem cells (CySCs) generate cyst cells, which encapsulate the germline to support its development. We find that CySCs are maintained independently of niche self-renewal signals if activity of the PI3K/Tor pathway is inhibited. Conversely, PI3K/Tor is not sufficient alone to drive differentiation, suggesting that it acts to license cells for differentiation. Indeed, we find that the germline is required for differentiation of CySCs in response to PI3K/Tor elevation, indicating that final commitment to differentiation involves several steps and intercellular communication. We propose that CySC daughter cells are plastic, that their fate depends on the availability of neighbouring germ cells, and that PI3K/Tor acts to induce a primed state for CySC daughters to enable coordinated differentiation with the germline.


Adult Stem Cells/cytology , Drosophila Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , TOR Serine-Threonine Kinases/genetics , Testis/growth & development , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Germ Cells/growth & development , Male , Signal Transduction/genetics , Stem Cell Niche/genetics , Testis/metabolism
8.
Int J Mol Sci ; 22(24)2021 Dec 07.
Article En | MEDLINE | ID: mdl-34947977

Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research.


Adult Stem Cells/cytology , Organ Culture Techniques/methods , Organoids/cytology , Cell Differentiation , Heart/physiology , Humans , Models, Biological , Pluripotent Stem Cells/cytology , Regeneration , Spheroids, Cellular/cytology
9.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article En | MEDLINE | ID: mdl-34768825

Heart failure pathobiology is permissive to reparative intent. Regenerative therapies exemplify an emerging disruptive innovation aimed at achieving structural and functional organ restitution. However, mixed outcomes, complexity in use, and unsustainable cost have curtailed broader adoption, mandating the development of novel cardio-regenerative approaches. Lineage guidance offers a standardized path to customize stem cell fitness for therapy. A case in point is the molecular induction of the cardiopoiesis program in adult stem cells to yield cardiopoietic cell derivatives designed for heart failure treatment. Tested in early and advanced clinical trials in patients with ischemic heart failure, clinical grade cardiopoietic cells were safe and revealed therapeutic improvement within a window of treatment intensity and pre-treatment disease severity. With the prospect of mass customization, cardiopoietic guidance has been streamlined from the demanding, recombinant protein cocktail-based to a protein-free, messenger RNA-based single gene protocol to engineer affordable cardiac repair competent cells. Clinical trial biobanked stem cells enabled a systems biology deconvolution of the cardiopoietic cell secretome linked to therapeutic benefit, exposing a paracrine mode of action. Collectively, this new knowledge informs next generation regenerative therapeutics manufactured as engineered cellular or secretome mimicking cell-free platforms. Launching biotherapeutics tailored for optimal outcome and offered at mass production cost would contribute to advancing equitable regenerative care that addresses population health needs.


Heart Failure/rehabilitation , Heart Failure/therapy , Regenerative Medicine/methods , Adult Stem Cells/cytology , Humans , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Stem Cells/cytology
10.
Cells ; 10(11)2021 11 04.
Article En | MEDLINE | ID: mdl-34831242

The regulation of adult neural stem or progenitor cell (aNSC) proliferation and differentiation as an interplay of cell-intrinsic and local environmental cues remains in part unclear, impeding their role in putative regenerative therapies. aNSCs with all major properties of NSCs in vitro have been identified in a variety of brain regions beyond the classic neurogenic niches, including the caudal periventricular regions (PVRs) of the midbrain, though active neurogenesis is either limited or merely absent in these regions. To elucidate cell-intrinsic properties of aNSCs from various PVRs, we here examined the proliferation and early differentiation capacity of murine aNSCs from non-neurogenic midbrain PVRs (PVRMB) compared to aNSCs from the neurogenic ventricular-subventricular zone (PVRV-SVZ) 7 days after transplantation into the permissive pro-neurogenic niche of the dentate gyrus (DG) of the hippocampus in mice. An initial in vitro characterization of the transplants displayed very similar characteristics of both aNSC grafts after in vitro expansion with equal capacities of terminal differentiation into astrocytes and Tuj1+ neurons. Upon the allogenic transplantation of the respective aNSCs into the DG, PVRMB grafts showed a significantly lower graft survival and proliferative capacity compared to PVRV-SVZ transplants, whereby the latter are exclusively capable of generating new neurons. Although these differences might be-in part-related to the transplantation procedure and the short-term study design, our data strongly imply important cell-intrinsic differences between aNSCs from neurogenic compared to non-neurogenic PVRs with respect to their neurogenic potential and/or their sensitivity to neurogenic cues.


Adult Stem Cells/cytology , Hippocampus/cytology , Mesencephalon/cytology , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurogenesis , Stem Cell Niche , Animals , Cell Differentiation , Cell Proliferation , Graft Survival , Mice, Inbred C57BL , Mice, Transgenic , Physical Conditioning, Animal , SOXB1 Transcription Factors/metabolism
11.
Sci Rep ; 11(1): 21013, 2021 10 25.
Article En | MEDLINE | ID: mdl-34697387

PAX6 is a highly conserved transcription factor and key regulator of several neurogenic processes, including the continuous generation of dopaminergic/GABAergic interneurons in the adult ventricular-subventricular (V-SVZ) neurogenic system in mice. Here we report that PAX6 cooperates with the TALE-homeodomain transcription factor PBX1 in this context. Chromatin-immunoprecipitation showed that PBX1 and PAX6 co-occupy shared genomic binding sites in adult V-SVZ stem- and progenitor cell cultures and mouse embryonic stem cells, while depletion of Pbx1 revealed that association of PAX6 with these sites requires the presence of PBX1. Expression profiling together with viral overexpression or knockdown of Pax6 or Pbx1 identified novel PBX1-PAX6 co-regulated genes, including several transcription factors. Computational modeling of genome wide expression identified novel cross-regulatory networks among these very transcription factors. Taken together, the results presented here highlight the intimate link that exists between PAX6 and TALE-HD family proteins and contribute novel insights into how the orchestrated activity of transcription factors shapes adult V-SVZ neurogenesis.


Epistasis, Genetic , Gene Expression Regulation , Neural Stem Cells/metabolism , PAX6 Transcription Factor/genetics , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Computational Biology/methods , Female , Gene Expression Profiling , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Neural Stem Cells/cytology , Neurogenesis/genetics , PAX6 Transcription Factor/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Binding
12.
Biol Open ; 10(10)2021 10 15.
Article En | MEDLINE | ID: mdl-34648017

In vitro production of tissue-specific stem cells [e.g. haematopoietic stem cells (HSCs)] is a key goal of regenerative medicine. However, recent efforts to produce fully functional tissue-specific stem cells have fallen short. One possible cause of shortcomings may be that model organisms used to characterize basic vertebrate embryology (Xenopus, zebrafish, chick) may employ molecular mechanisms for stem cell specification that are not conserved in humans, a prominent example being the specification of primordial germ cells (PGCs). Germ plasm irreversibly specifies PGCs in many models; however, it is not conserved in humans, which produce PGCs from tissue termed germline-competent mesoderm (GLCM). GLCM is not conserved in organisms containing germ plasm, or even in mice, but understanding its developmental potential could unlock successful production of other stem cell types. GLCM was first discovered in embryos from the axolotl and its conservation has since been demonstrated in pigs, which develop from a flat-disc embryo like humans. Together these findings suggest that GLCM is a conserved basal trait of vertebrate embryos. Moreover, the immortal nature of germ cells suggests that immortality is retained during GLCM specification; here we suggest that the demonstrated pluripotency of GLCM accounts for retention of immortality in somatic stem cell types as well. This article has an associated Future Leaders to Watch interview with the author of the paper.


Adult Stem Cells/cytology , Embryo, Mammalian/embryology , Embryo, Nonmammalian/embryology , Germ Cells/growth & development , Mesoderm/embryology , Animals , Chick Embryo , Mice , Swine , Xenopus , Zebrafish
13.
Genes (Basel) ; 12(10)2021 09 24.
Article En | MEDLINE | ID: mdl-34680880

Populations of resident stem cells (SCs) are responsible for maintaining, repairing, and regenerating adult tissues. In addition to having the capacity to generate all the differentiated cell types of the tissue, adult SCs undergo long periods of quiescence within the niche to maintain themselves. The process of SC renewal and differentiation is tightly regulated for proper tissue regeneration throughout an organisms' lifetime. Epigenetic regulators, such as the polycomb group (PcG) of proteins have been implicated in modulating gene expression in adult SCs to maintain homeostatic and regenerative balances in adult tissues. In this review, we summarize the recent findings that elucidate the composition and function of the polycomb repressive complex machinery and highlight their role in diverse adult stem cell compartments.


Epigenesis, Genetic/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Polycomb-Group Proteins/genetics , Adult Stem Cells/cytology , Cell Differentiation/genetics , Cell Self Renewal/genetics , Humans
14.
Front Immunol ; 12: 716606, 2021.
Article En | MEDLINE | ID: mdl-34539651

Recent clinical experience has demonstrated that adoptive regulatory T (Treg) cell therapy is a safe and feasible strategy to suppress immunopathology via induction of host tolerance to allo- and autoantigens. However, clinical trials continue to be compromised due to an inability to manufacture a sufficient Treg cell dose. Multipotent adult progenitor cells (MAPCⓇ) promote Treg cell differentiation in vitro, suggesting they may be repurposed to enhance ex vivo expansion of Tregs for adoptive cellular therapy. Here, we use a Good Manufacturing Practice (GMP) compatible Treg expansion platform to demonstrate that MAPC cell-co-cultured Tregs (MulTreg) exhibit a log-fold increase in yield across two independent cohorts, reducing time to target dose by an average of 30%. Enhanced expansion is coupled to a distinct Treg cell-intrinsic transcriptional program characterized by elevated expression of replication-related genes (CDK1, PLK1, CDC20), downregulation of progenitor and lymph node-homing molecules (LEF1 CCR7, SELL) and induction of intestinal and inflammatory tissue migratory markers (ITGA4, CXCR1) consistent with expression of a gut homing (CCR7lo ß7hi) phenotype. Importantly, we find that MulTreg are more readily expanded from patients with autoimmune disease compared to matched Treg lines, suggesting clinical utility in gut and/or T helper type1 (Th1)-driven pathology associated with autoimmunity or transplantation. Relative to expanded Tregs, MulTreg retain equivalent and robust purity, FoxP3 Treg-Specific Demethylated Region (TSDR) demethylation, nominal effector cytokine production and potent suppression of Th1-driven antigen specific and polyclonal responses in vitro and xeno Graft vs Host Disease (xGvHD) in vivo. These data support the use of MAPC cell co-culture in adoptive Treg therapy platforms as a means to rescue expansion failure and reduce the time required to manufacture a stable, potently suppressive product.


Autoimmunity , Lymphocyte Count , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/immunology , Adult Stem Cells/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Biomarkers , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Regulation , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Humans , Immunophenotyping , Male , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
Mol Pharm ; 18(11): 3931-3950, 2021 11 01.
Article En | MEDLINE | ID: mdl-34582198

Organoids have complex three-dimensional structures that exhibit functionalities and feature architectures similar to those of in vivo organs and are developed from adult stem cells, embryonic stem cells, and pluripotent stem cells through a self-organization process. Organoids derived from adult epithelial stem cells are the most mature and extensive. In recent years, using organoid culture techniques, researchers have established various adult human tissue-derived epithelial organoids, including intestinal, colon, lung, liver, stomach, breast, and oral mucosal organoids, all of which exhibit strong research and application prospects. Studies have shown that epithelial organoids are mainly applied in drug discovery, personalized drug response testing, disease mechanism research, and regenerative medicine. In this review, we mainly discuss current organoid culture systems and potential applications of this technique with human epithelial tissue.


Adult Stem Cells/cytology , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , Epithelial Cells/cytology , Organoids , Cell Culture Techniques/trends , Drug Evaluation, Preclinical/trends , Humans
16.
Cells ; 10(8)2021 08 10.
Article En | MEDLINE | ID: mdl-34440811

NF-κB (nuclear factor kappa B) belongs to a family of transcription factors known to regulate a broad range of processes such as immune cell function, proliferation and cancer, neuroprotection, and long-term memory. Upcoming fields of NF-κB research include its role in stem cells and developmental processes. In the present review, we discuss one role of NF-κB in development in Drosophila, Xenopus, mice, and humans in accordance with the concept of evo-devo (evolutionary developmental biology). REL domain-containing proteins of the NF-κB family are evolutionarily conserved among these species. In addition, we summarize cellular phenotypes such as defective B- and T-cell compartments related to genetic NF-κB defects detected among different species. While NF-κB proteins are present in nearly all differentiated cell types, mouse and human embryonic stem cells do not contain NF-κB proteins, potentially due to miRNA-dependent inhibition. However, the mesodermal and neuroectodermal differentiation of mouse and human embryonic stem cells is hampered upon the repression of NF-κB. We further discuss NF-κB as a crucial regulator of differentiation in adult stem cells such as neural crest-derived and mesenchymal stem cells. In particular, c-REL seems to be important for neuronal differentiation and the neuroprotection of human adult stem cells, while RELA plays a crucial role in osteogenic and mesodermal differentiation.


NF-kappa B/metabolism , Stem Cells/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , MicroRNAs/metabolism , NF-kappa B/deficiency , NF-kappa B/genetics , Signal Transduction , Stem Cells/cytology , Transcription Factor RelA/metabolism
17.
Cells ; 10(8)2021 08 10.
Article En | MEDLINE | ID: mdl-34440814

Adult neural stem and progenitor cells (NSPCs) contribute to learning, memory, maintenance of homeostasis, energy metabolism and many other essential processes. They are highly heterogeneous populations that require input from a regionally distinct microenvironment including a mix of neurons, oligodendrocytes, astrocytes, ependymal cells, NG2+ glia, vasculature, cerebrospinal fluid (CSF), and others. The diversity of NSPCs is present in all three major parts of the CNS, i.e., the brain, spinal cord, and retina. Intrinsic and extrinsic signals, e.g., neurotrophic and growth factors, master transcription factors, and mechanical properties of the extracellular matrix (ECM), collectively regulate activities and characteristics of NSPCs: quiescence/survival, proliferation, migration, differentiation, and integration. This review discusses the heterogeneous NSPC populations in the normal physiology and highlights their potentials and roles in injured/diseased states for regenerative medicine.


Adult Stem Cells/physiology , Neural Stem Cells/physiology , Neurodegenerative Diseases/pathology , Spinal Cord Injuries/pathology , Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Animals , Antigens/metabolism , Cell Differentiation , Ependyma/cytology , Ependyma/physiology , Humans , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurodegenerative Diseases/therapy , Proteoglycans/metabolism , Regenerative Medicine , Spinal Cord Injuries/therapy
18.
Cells ; 10(8)2021 08 17.
Article En | MEDLINE | ID: mdl-34440881

Muscle stem cells (MuSCs) are essential for muscle growth, maintenance and repair. Over the past decade, experiments in Drosophila have been instrumental in understanding the molecular and cellular mechanisms regulating MuSCs (also known as adult muscle precursors, AMPs) during development. A large number of genetic tools available in fruit flies provides an ideal framework to address new questions which could not be addressed with other model organisms. This review reports the main findings revealed by the study of Drosophila AMPs, with a specific focus on how AMPs are specified and properly positioned, how they acquire their identity and which are the environmental cues controlling their behavior and fate. The review also describes the recent identification of the Drosophila adult MuSCs that have similar characteristics to vertebrates MuSCs. Integration of the different levels of MuSCs analysis in flies is likely to provide new fundamental knowledge in muscle stem cell biology largely applicable to other systems.


Muscle Development/physiology , Myoblasts/physiology , Regeneration/physiology , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Cell Differentiation , Cell Proliferation , Drosophila/cytology , Drosophila/physiology , Models, Biological , Myoblasts/cytology , Signal Transduction
19.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article En | MEDLINE | ID: mdl-34210091

(1) Background: We established a new bladder ischemia rat model through bilateral partial iliac arterial occlusion (BPAO) and investigated the therapeutic effect of adipose-derived stem cells (ADSCs) and ADSC-derived microvesicles (MVs); (2) Methods: The study included four groups: (1) sham, (2) BPAO, (3) BPAO + ADSCs, and (4) BPAO + ADSC-derived MVs. Female Wistar rats with BPAO were injected with ADSCs or ADSC-derived MVs through the femoral artery. Doppler flowmetry and real-time laser speckle contrast imaging were performed to quantify blood flow in the common iliac arteries and bladder microcirculation. A 24-h behavior study and transcystometrogram were conducted after 2 weeks. Bladder histology, immunostaining, and lipid peroxidation assay were performed. The expressions of P2X2, P2X3, M2, and M3 receptors and nerve growth factor (NGF) were evaluated; (3) Results: BPAO significantly reduced bladder microcirculation, intercontraction interval (ICI), and bladder volume and increased the amplitude of nonvoiding contraction, neutrophil infiltration, and malondialdehyde and NGF levels. ADSCs and ADSC-derived MVs significantly ameliorated these effects. The results of Western blot showed that the BPAO group exhibited the highest expression of M3 and P2X2 receptors. ADSCs significantly attenuated the expressions of M2 and P2X2 receptors. ADSC-derived MVs significantly attenuated the expressions of M3 and P2X2 receptors; (4) Conclusions: ADSCs and ADSC-derived MVs ameliorated the adverse effects of BPAO including bladder overactivity, bladder ischemia, and oxidative stress. Inflammation, muscarinic signaling, purinergic signaling, and NGF might be involved in the therapeutic mechanism.


Adipose Tissue/cytology , Adult Stem Cells/transplantation , Cell-Derived Microparticles/transplantation , Urinary Bladder, Overactive/therapy , Adult Stem Cells/cytology , Animals , Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/therapy , Cell-Derived Microparticles/physiology , Disease Models, Animal , Female , Iliac Artery/pathology , Ischemia/etiology , Ischemia/therapy , Rats , Rats, Wistar , Urinary Bladder/pathology , Urinary Bladder, Overactive/etiology
20.
Life Sci Alliance ; 4(9)2021 09.
Article En | MEDLINE | ID: mdl-34226276

DNA repair proteins are critical to the maintenance of genomic integrity. Specific types of genotoxic factors, including reactive oxygen species generated during normal cellular metabolism or as a result of exposure to exogenous oxidative agents, frequently leads to "ragged" single-strand DNA breaks. The latter exhibits abnormal free DNA ends containing either a 5'-hydroxyl or 3'-phosphate requiring correction by the dual function enzyme, polynucleotide kinase phosphatase (PNKP), before DNA polymerase and ligation reactions can occur to seal the break. Pnkp gene deletion during early murine development leads to lethality; in contrast, the role of PNKP in adult mice is unknown. To investigate the latter, we used an inducible conditional mutagenesis approach to cause global disruption of the Pnkp gene in adult mice. This resulted in a premature aging-like phenotype, characterized by impaired growth of hair follicles, seminiferous tubules, and neural progenitor cell populations. These results point to an important role for PNKP in maintaining the normal growth and survival of these murine progenitor populations.


Cell Self Renewal/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Stem Cells/cytology , Stem Cells/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Apoptosis , Biomarkers , Cell Differentiation/genetics , DNA Damage , DNA Repair , Dermis/cytology , Dermis/metabolism , Fluorescent Antibody Technique , Germ Cells/cytology , Germ Cells/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Hyperpigmentation/genetics , Immunohistochemistry , Melanins/metabolism , Mice , Mice, Knockout
...