Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39.439
Filter
1.
J Vis Exp ; (209)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39141532

ABSTRACT

Due to the limited accessibility of the human retina, retinal organoids (ROs) are the best model for studying human retinal disease, which could reveal the mechanism of retinal development and the occurrence of retinal disease. Microglia (MG) are unique resident macrophages in the retina and central nervous system (CNS), serving crucial immunity functions. However, retinal organoids lack microglia since their differentiation origin is the yolk sac. The specific pathogenesis of microglia in these retinal diseases remains unclear; therefore, the establishment of a microglia-incorporated retinal organoid model turns out to be necessary. Here, we successfully constructed a co-cultured model of retinal organoids with microglia derived from human stem cells. In this article, we differentiated microglia and then co-cultured to retinal organoids in the early stage. As the incorporation of immune cells, this model provides an optimized platform for retinal disease modeling and drug screening to facilitate in-depth research on the pathogenesis and treatment of retinal and CNS-related diseases.


Subject(s)
Coculture Techniques , Microglia , Organoids , Retina , Organoids/cytology , Microglia/cytology , Retina/cytology , Humans , Coculture Techniques/methods , Cell Differentiation/physiology
2.
Development ; 151(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38950937

ABSTRACT

The capacity to regenerate lost tissues varies significantly among animals. Some phyla, such as the annelids, display substantial regenerating abilities, although little is known about the cellular mechanisms underlying the process. To precisely determine the origin, plasticity and fate of the cells participating in blastema formation and posterior end regeneration after amputation in the annelid Platynereis dumerilii, we developed specific tools to track different cell populations. Using these tools, we find that regeneration is partly promoted by a population of proliferative gut cells whose regenerative potential varies as a function of their position along the antero-posterior axis of the worm. Gut progenitors from anterior differentiated tissues are lineage restricted, whereas gut progenitors from the less differentiated and more proliferative posterior tissues are much more plastic. However, they are unable to regenerate the stem cells responsible for the growth of the worms. Those stem cells are of local origin, deriving from the cells present in the segment abutting the amputation plane, as are most of the blastema cells. Our results favour a hybrid and flexible cellular model for posterior regeneration in Platynereis relying on different degrees of cell plasticity.


Subject(s)
Cell Plasticity , Cell Proliferation , Polychaeta , Regeneration , Animals , Regeneration/physiology , Polychaeta/physiology , Polychaeta/cytology , Cell Plasticity/physiology , Stem Cells/cytology , Cell Differentiation/physiology , Annelida/physiology
4.
Mech Ageing Dev ; 220: 111959, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950628

ABSTRACT

Oligodendrocyte precursor cells (OPCs) comprise 5-8 % of the adult glial cell population and stand out as the most proliferative cell type in the central nervous system (CNS). OPCs are responsible for generating oligodendrocytes (OLs), the myelinating cells of the CNS. However, OPC functions decline as we age, resulting in impaired differentiation and inadequate remyelination. This review explores the cellular and molecular changes associated with OPC aging, and their impact on OPC differentiation and functionality. Furthermore, it examines the impact of OPC aging within the context of multiple sclerosis and Alzheimer's disease, both neurodegenerative conditions wherein aged OPCs exacerbate disease progression by impeding remyelination. Moreover, various pharmacological interventions targeting pathways related to senescence and differentiation are discussed as potential strategies to rejuvenate aged OPCs. Enhancing our understanding of OPC aging mechanisms holds promise for developing new therapies to improve remyelination and repair in age-related neurodegenerative disorders.


Subject(s)
Brain , Cell Differentiation , Cellular Senescence , Oligodendrocyte Precursor Cells , Humans , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/physiology , Cellular Senescence/physiology , Animals , Brain/metabolism , Brain/pathology , Cell Differentiation/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Aging/physiology , Aging/metabolism , Aging/pathology , Oligodendroglia/metabolism , Remyelination/physiology
5.
Bull Exp Biol Med ; 177(1): 35-38, 2024 May.
Article in English | MEDLINE | ID: mdl-38954301

ABSTRACT

The features of the participation of Smad3 in the functioning of neural stem cells (NSC), neuronal committed precursors (NCP), and neuroglial elements were studied in vitro. It was found that this intracellular signaling molecule enhances the clonogenic and proliferative activities of NCP and inhibits specialization of neuronal precursors. At the same time, Smad3 does not participate in the realization of the growth potential of NSC. With regard to the secretory function (production of neurotrophic growth factors) of neuroglial cells, the stimulating role of Smad3-mediated signaling was shown. These results indicate the promise of studying the possibility of using Smad3 as a fundamentally new target for neuroregenerative agents.


Subject(s)
Cell Proliferation , Neural Stem Cells , Neuroglia , Smad3 Protein , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Animals , Neuroglia/metabolism , Neuroglia/cytology , Cell Proliferation/physiology , Signal Transduction , Cell Differentiation/physiology , Cells, Cultured , Rats , Neurons/metabolism , Neurons/cytology , Mice
6.
Mol Neurodegener ; 19(1): 53, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997755

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY: In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION: We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.


Subject(s)
Inflammation , Multiple Sclerosis , Oligodendrocyte Precursor Cells , Remyelination , Humans , Remyelination/physiology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Multiple Sclerosis/pathology , Animals , Inflammation/immunology , Cell Differentiation/physiology , Myelin Sheath , Oligodendroglia
7.
Respir Res ; 25(1): 277, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010108

ABSTRACT

To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.


Subject(s)
Induced Pluripotent Stem Cells , Lung , Respiratory Tract Infections , Virus Diseases , Humans , Lung/immunology , Lung/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Animals , Cell Differentiation/physiology , Models, Biological
8.
J Orthop Surg Res ; 19(1): 407, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014435

ABSTRACT

PURPOSE: Oncostatin M (OSM) is involved in the regulation of osteogenic differentiation and has a major role in the development of heterotopic ossification. The role of OSM in osteogenic differentiation of tendon-derived stem cells (TDSCs) and its mechanism have not been reported. This study aim to investigate the role of OSM in osteogenic differentiation of TDSCs and study the mechanism. METHODS: TDSCs were differentiated in osteogenic differentiation medium for 7 days. Recombinant OSM was added to the osteogenic differentiation medium for 7 and 14 days. The effect of Janus kinase 2 (JAK2) inhibitor AZD1480 and signal transducer and activator of transcription 3 (STAT3) inhibitor stattic in the presence of recombinant OSM on osteogenic differentiation of TDSCs was examined after differentiation for 7 and 14 days. Alkaline phosphatase and alizarin red staining were used to assess the effects on early and mid-stage osteogenic differentiation, respectively. Western blotting and qPCR were used to assess the expression of receptor and signalling pathway-related proteins and osteogenic marker genes, respectively. RESULTS: TDSCs were successfully induced to differentiate into osteoblasts. Recombinant OSM promoted osteogenic differentiation of TDSCs to early and mid-stages. After addition of AZD1480 or stattic, decreased alkaline phosphatase and alizarin red staining were observed in the early and mid-stages of osteogenic differentiation. Additionally, decreased expression of receptor and pathway-related proteins, and osteogenic genes was found by western blotting and qPCR, respectively. CONCLUSION: OSM promotes osteogenic differentiation of TDSCs and the JAK2/STAT3 signalling pathway plays an important role.


Subject(s)
Cell Differentiation , Janus Kinase 2 , Oncostatin M , Osteogenesis , STAT3 Transcription Factor , Signal Transduction , Stem Cells , Tendons , Oncostatin M/pharmacology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Osteogenesis/drug effects , Osteogenesis/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Tendons/cytology , Stem Cells/drug effects , Humans , Cells, Cultured , Animals
9.
ASN Neuro ; 16(1): 2371163, 2024.
Article in English | MEDLINE | ID: mdl-39024549

ABSTRACT

The fifteen canonical paracrine fibroblast growth factors (FGFs) are organized in five subfamilies that interact with four FGF-receptors (FGFRs) and heparan sulfate proteoglycan (HSPG) co-receptors. Many of these FGFs are expressed in CNS regions where oligodendrocyte (OL) progenitors originate, migrate or differentiate. FGF2 (basic FGF) is considered a prototype FGF and the information about the effects of FGF signaling on OL-lineage cells has evolved largely from the study of FGF2. However, other FGFs from four subfamilies ((FGF1 (FGF1,-2), FGF4 (FGF4,-5,-6), FGF8 (FGF8,-17,-18) and FGF9 (FGF9,-16,-20)) that can interact with the isoforms of FGFRs expressed in OL-lineage cells may also play important roles. We previously reported OL-responses to FGF8 family members. Here, we investigate the effects of members of the FGF1,-4, and -9 subfamilies on proliferation and differentiation of OL progenitors (OPCs), and on cell cycle re-entry and down-regulation of myelin proteins by mature OLs. We found that while FGF2 induced all these responses strongly, FGF4,-6,-9 could do so only transiently and in the presence of exogenous HSPGs, and that FGF5,-16,-20 could not do so even in the presence of heparin or at higher concentrations. Furthermore, we noted that structurally similar FGFs within subfamilies did not always show similarities in their biological effects on OL-lineage cells. Taken together, these studies reveal that FGFs differ in the way they regulate the OL-lineage cells, emphasizes the selectivity and importance of HSPGs as FGF co-receptors in OL-lineage cells and suggests that structural similarity among FGF-subfamily members may not always predict their overlapping biological functions.


Structurally similar members of the FGF1, -4, and -9 subfamilies trigger diverse biological responses in oligodendrocyte-lineage cells and exhibit selective requirement for heparan sulfate proteoglycans as FGF co-receptors.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Oligodendroglia , Animals , Oligodendroglia/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Cell Differentiation/physiology , Cell Differentiation/drug effects , Cell Proliferation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Structure-Activity Relationship , Rats
10.
Cell Syst ; 15(7): 649-661.e9, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38981488

ABSTRACT

Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.


Subject(s)
Organoids , Organoids/cytology , Organoids/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nephrons/cytology , Cell Differentiation/physiology , Stem Cells/cytology
11.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-39009447

ABSTRACT

Generation of human induced pluripotent stem cell (hiPSC)-derived motor neurons (MNs) offers an unprecedented approach to modeling movement disorders such as dystonia and amyotrophic lateral sclerosis. However, achieving survival poses a significant challenge when culturing induced MNs, especially when aiming to reach late maturation stages. Utilizing hiPSC-derived motor neurons and primary mouse astrocytes, we assembled two types of coculture systems: direct coculturing of neurons with astrocytes and indirect coculture using culture inserts that physically separate neurons and astrocytes. Both systems significantly enhance neuron survival. Compared with these two systems, no significant differences in neurodevelopment, maturation, and survival within 3 weeks, allowing to prepare neurons at maturation stages. Using the indirect coculture system, we obtained highly pure MNs at the late mature stage from hiPSCs. Transcriptomic studies of hiPSC-derived MNs showed a typical neurodevelopmental switch in gene expression from the early immature stage to late maturation stages. Mature genes associated with neurodevelopment and synaptogenesis are highly enriched in MNs at late stages, demonstrating that these neurons achieve maturation. This study introduces a novel tool for the preparation of highly pure hiPSC-derived neurons, enabling the determination of neurological disease pathogenesis in neurons at late disease onset stages through biochemical approaches, which typically necessitate highly pure neurons. This advancement is particularly significant in modeling age-related neurodegeneration.


Subject(s)
Astrocytes , Coculture Techniques , Induced Pluripotent Stem Cells , Motor Neurons , Induced Pluripotent Stem Cells/physiology , Animals , Motor Neurons/physiology , Mice , Astrocytes/physiology , Humans , Cell Differentiation/physiology , Cells, Cultured , Neurogenesis/physiology
12.
Nat Rev Neurosci ; 25(8): 519-534, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951687

ABSTRACT

During central nervous system (CNS) development, neural progenitor cells (NPCs) generate neurons and glia in two different ways. In direct neurogenesis, daughter cells differentiate directly into neurons or glia, whereas in indirect neurogenesis, neurons or glia are generated after one or more daughter cell divisions. Intriguingly, indirect neurogenesis is not stochastically deployed and plays instructive roles during CNS development: increased generation of cells from specific lineages; increased generation of early or late-born cell types within a lineage; and increased cell diversification. Increased indirect neurogenesis might contribute to the anterior CNS expansion evident throughout the Bilateria and help to modify brain-region size without requiring increased NPC numbers or extended neurogenesis. Increased indirect neurogenesis could be an evolutionary driver of the gyrencephalic (that is, folded) cortex that emerged during mammalian evolution and might even have increased during hominid evolution. Thus, selection of indirect versus direct neurogenesis provides a powerful developmental and evolutionary instrument that drives not only the evolution of CNS complexity but also brain expansion and modulation of brain-region size, and thereby the evolution of increasingly advanced cognitive abilities. This Review describes indirect neurogenesis in several model species and humans, and highlights some of the molecular genetic mechanisms that control this important process.


Subject(s)
Neurogenesis , Neurogenesis/physiology , Humans , Animals , Biological Evolution , Neural Stem Cells/physiology , Neural Stem Cells/cytology , Neurons/physiology , Cell Differentiation/physiology , Central Nervous System/physiology , Central Nervous System/growth & development , Central Nervous System/cytology , Neuroglia/physiology , Brain/physiology , Brain/growth & development , Brain/cytology
13.
J Orthop Surg Res ; 19(1): 386, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951811

ABSTRACT

BACKGROUND: Bone defects, resulting from substantial bone loss that exceeds the natural self-healing capacity, pose significant challenges to current therapeutic approaches due to various limitations. In the quest for alternative therapeutic strategies, bone tissue engineering has emerged as a promising avenue. Notably, excretory proteins from Toxoplasma gondii (TgEP), recognized for their immunogenicity and broad spectrum of biological activities secreted or excreted during the parasite's lifecycle, have been identified as potential facilitators of osteogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Building on our previous findings that TgEP can enhance osteogenic differentiation, this study investigated the molecular mechanisms underlying this effect and assessed its therapeutic potential in vivo. METHODS: We determined the optimum concentration of TgEP through cell cytotoxicity and cell proliferation assays. Subsequently, hBMSCs were treated with the appropriate concentration of TgEP. We assessed osteogenic protein markers, including alkaline phosphatase (ALP), Runx2, and Osx, as well as components of the BMP/Smad signaling pathway using quantitative real-time PCR (qRT-PCR), siRNA interference of hBMSCs, Western blot analysis, and other methods. Furthermore, we created a bone defect model in Sprague-Dawley (SD) male rats and filled the defect areas with the GelMa hydrogel, with or without TgEP. Microcomputed tomography (micro-CT) was employed to analyze the bone parameters of defect sites. H&E, Masson and immunohistochemical staining were used to assess the repair conditions of the defect area. RESULTS: Our results indicate that TgEP promotes the expression of key osteogenic markers, including ALP, Runx2, and Osx, as well as the activation of Smad1, BMP2, and phosphorylated Smad1/5-crucial elements of the BMP/Smad signaling pathway. Furthermore, in vivo experiments using a bone defect model in rats demonstrated that TgEP markedly promoted bone defect repair. CONCLUSION: Our results provide compelling evidence that TgEP facilitates hBMSC osteogenic differentiation through the BMP/Smad signaling pathway, highlighting its potential as a therapeutic approach for bone tissue engineering for bone defect healing.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Rats, Sprague-Dawley , Signal Transduction , Toxoplasma , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Humans , Animals , Signal Transduction/physiology , Cell Differentiation/physiology , Male , Toxoplasma/physiology , Rats , Smad Proteins/metabolism , Protozoan Proteins/metabolism , Bone Morphogenetic Proteins/metabolism , Cells, Cultured
14.
J Orthop Surg Res ; 19(1): 396, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982418

ABSTRACT

BACKGROUND: The progression of osteoporosis (OP) can dramatically increase the risk of fractures, which seriously disturb the life of elderly individuals. Specific protein 1 (SP1) is involved in OP progression. However, the mechanism by which SP1 regulates OP progression remains unclear. OBJECTIVE: This study investigated the mechanism underlying the function of SP1 in OP. METHODS: SAMP6 mice were used to establish an in vivo model of age-dependent OP, and BALB/c mice were used as controls. BMSCs were extracted from two subtypes of mice. Hematoxylin and eosin staining were performed to mark the intramedullary trabecular bone structure to evaluate histological changes. ChIP assay was used to assess the targeted regulation between SP1 and miR-133a-3p. The binding sites between MAPK3 and miR-133a-3p were verified using a dual-luciferase reporter assay. The mRNA levels of miR-133a-3p and MAPK3 were detected using quantitative reverse transcription polymerase chain reaction (RT-qPCR). The protein expression of SP1, MAPK3, Colla1, OCN, and Runx2 was examined using Western blotting. Alkaline phosphatase (ALP) kit and Alizarin Red S staining were used to investigate ALP activity and mineralized nodules, respectively. RESULTS: The levels of SP1 and miR-133a-3p were upregulated, whereas the expression of MAPK3 was downregulated in BMSCs from SAMP6 mice, and miR-133a-3p inhibitor accelerated osteogenic differentiation in BMSCs. SP1 directly targeted miR-133a-3p, and MAPK3 was the downstream mRNA of miR-133a-3p. Mechanically, SP1 accelerated osteogenic differentiation in BMSCs via transcriptional mediation of the miR-133a-3p/MAPK3 axis. CONCLUSION: SP1 regulates osteogenic differentiation by mediating the miR-133a-3p/MAPK3 axis, which would shed new light on strategies for treating senile OP.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , MicroRNAs , Mitogen-Activated Protein Kinase 3 , Osteogenesis , Osteoporosis , Sp1 Transcription Factor , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mice, Inbred BALB C , Cells, Cultured , Disease Models, Animal , Male
15.
J Vis Exp ; (209)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39072637

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is largely attributed to tobacco smoke exposure. Investigating how airway epithelial cells functionally adapt to tobacco smoke is crucial for understanding the pathogenesis of COPD. The present study was to set up an in vitro model using primary murine airway epithelial cells to mimic the real-life impact of tobacco smoke. Unlike established cell lines, primary cells retain more in vivo-like properties, including growth patterns, aging, and differentiation. These cells exhibit a sensitive inflammatory response and efficient differentiation, thus closely representing physiological conditions. In this model, primary murine airway epithelial cells were cultured for 28 days under an air-liquid interface with an optimal concentration of cigarette smoke extract (CSE), which led to the transformation of a monolayer of undifferentiated cells into a pseudostratified columnar epithelium, indicative of CSE acclimation. Comprehensive multi-omics analyses were then applied to elucidate the mechanisms by which CSE influences the differentiation of basal airway cells. These insights provide a deeper understanding of the cellular processes underpinning COPD progression in response to tobacco smoke exposure.


Subject(s)
Cell Differentiation , Epithelial Cells , Smoke , Animals , Mice , Cell Differentiation/drug effects , Cell Differentiation/physiology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Smoke/adverse effects , Nicotiana/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Tobacco Products , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/metabolism , Multiomics
16.
Neuroscience ; 552: 142-151, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38960088

ABSTRACT

Hippocampus is a critical component of the central nervous system. SRSF10 is expressed in central nervous system and plays important roles in maintaining normal brain functions. However, its role in hippocampus development is unknown. In this study, using SRSF10 conditional knock-out mice in neural progenitor cells (NPCs), we found that dysfunction of SRSF10 leads to developmental defects in the dentate gyrus of hippocampus, which manifests as the reduced length and wider suprapyramidal blade and infrapyramidal blade.Furthermore, we proved that loss of SRSF10 in NPCs caused inhibition of the differentiation activity and the abnormal migration of NPCs and granule cells, resulting in reduced granule cells and more ectopic granule cells dispersed in the molecular layer and hilus. Finally, we found that the abnormal migration may be caused by the radial glia scaffold and the reduced DISC1 expression in NPCs. Together, our results indicate that SRSF10 is required for the cell migration and formation of dentate gyrus during the development of hippocampus.


Subject(s)
Cell Movement , Dentate Gyrus , Mice, Knockout , Neural Stem Cells , Serine-Arginine Splicing Factors , Animals , Neural Stem Cells/metabolism , Dentate Gyrus/metabolism , Cell Movement/physiology , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Neurons/metabolism , Hippocampus/metabolism , Mice , Neurogenesis/physiology , Cell Differentiation/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice, Inbred C57BL
17.
Neuropharmacology ; 257: 110058, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38960135

ABSTRACT

Postnatal hippocampal neurogenesis is essential for learning and memory. Hippocampal neural precursor cells (NPCs) can be induced to proliferate and differentiate into either glial cells or dentate granule cells. Notably, hippocampal neurogenesis decreases dramatically with age, partly due to a reduction in the NPC pool and a decrease in their proliferative activity. Alpha-melanocyte-stimulating hormone (α-MSH) improves learning, memory, neuronal survival and plasticity. Here, we used postnatally-isolated hippocampal NPCs from Wistar rat pups (male and female combined) to determine the role of the melanocortin analog [Nle4, D-Phe7]-α-MSH (NDP-MSH) in proliferation and fate acquisition of NPCs. Incubation of growth-factor deprived NPCs with 10 nM NDP-MSH for 6 days increased the proportion of Ki-67- and 5-bromo-2'-deoxyuridine (BrdU)-positive cells, compared to the control group, and these effects were blocked by the MC4R antagonist JKC-363. NDP-MSH also increased the proportion of glial fibrillar acidic protein (GFAP)/Ki-67, GFAP/sex-determining region Y-box2 (SOX2) and neuroepithelial stem cell protein (NESTIN)/Ki-67-double positive cells (type-1 and type-2 precursors). Finally, NDP-MSH induced peroxisome proliferator-activated receptor (PPAR)-γ protein expression, and co-incubation with the PPAR-γ inhibitor GW9662 prevented the effect of NDP-MSH on NPC proliferation and differentiation. Our results indicate that in vitro activation of MC4R in growth-factor-deprived postnatal hippocampal NPCs induces proliferation and promotes the relative expansion of the type-1 and type-2 NPC pool through a PPAR-γ-dependent mechanism. These results shed new light on the mechanisms underlying the beneficial effects of melanocortins in hippocampal plasticity and provide evidence linking the MC4R and PPAR-γ pathways in modulation of hippocampal NPC proliferation and differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Hippocampus , Neural Stem Cells , Neurogenesis , Rats, Wistar , Receptor, Melanocortin, Type 4 , alpha-MSH , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology , alpha-MSH/analogs & derivatives , Female , Cell Differentiation/drug effects , Cell Differentiation/physiology , Male , Neurogenesis/drug effects , Neurogenesis/physiology , Rats , Cells, Cultured , SOXB1 Transcription Factors/metabolism , Animals, Newborn , Glial Fibrillary Acidic Protein/metabolism , PPAR gamma/metabolism
18.
Math Med Biol ; 41(2): 135-155, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38970827

ABSTRACT

We discuss the mathematical modelling of two of the main mechanisms that pushed forward the emergence of multicellularity: phenotype divergence in cell differentiation and between-cell cooperation. In line with the atavistic theory of cancer, this disease being specific of multicellular animals, we set special emphasis on how both mechanisms appear to be reversed, however not totally impaired, rather hijacked, in tumour cell populations. Two settings are considered: the completely innovating, tinkering, situation of the emergence of multicellularity in the evolution of species, which we assume to be constrained by external pressure on the cell populations, and the completely planned-in the body plan-situation of the physiological construction of a developing multicellular animal from the zygote, or of bet hedging in tumours, assumed to be of clonal formation, although the body plan is largely-but not completely-lost in its constituting cells. We show how cancer impacts these two settings and we sketch mathematical models for them. We present here our contribution to the question at stake with a background from biology, from mathematics and from philosophy of science.


Subject(s)
Models, Biological , Neoplasms , Phenotype , Neoplasms/pathology , Neoplasms/physiopathology , Humans , Animals , Cell Differentiation/physiology , Mathematical Concepts , Cell Communication/physiology , Biological Evolution
19.
J Vis Exp ; (208)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38975788

ABSTRACT

Tendons and ligaments (T/L) are strong hierarchically organized structures uniting the musculoskeletal system. These tissues have a strictly arranged collagen type I-rich extracellular matrix (ECM) and T/L-lineage cells mainly positioned in parallel rows. After injury, T/L require a long time for rehabilitation with high failure risk and often unsatisfactory repair outcomes. Despite recent advancements in T/L biology research, one of the remaining challenges is that the T/L field still lacks a standardized differentiation protocol that is able to recapitulate T/L formation process in vitro. For example, bone and fat differentiation of mesenchymal precursor cells require just standard two-dimensional (2D) cell culture and the addition of specific stimulation media. For differentiation to cartilage, three-dimensional (3D) pellet culture and supplementation of TGFß is necessary. However, cell differentiation to tendon needs a very orderly 3D culture model, which ideally should also be subjectable to dynamic mechanical stimulation. We have established a 3-step (expansion, stimulation, and maturation) organoid model to form a 3D rod-like structure out of a self-assembled cell sheet, which delivers a natural microenvironment with its own ECM, autocrine, and paracrine factors. These rod-like organoids have a multi-layered cellular architecture within rich ECM and can be handled quite easily for exposure to static mechanical strain. Here, we demonstrated the 3-step protocol by using commercially available dermal fibroblasts. We could show that this cell type forms robust and ECM-abundant organoids. The described procedure can be further optimized in terms of culture media and optimized toward dynamic axial mechanical stimulation. In the same way, alternative cell sources can be tested for their potential to form T/L organoids and thus undergo T/L differentiation. In sum, the established 3D T/L organoid approach can be used as a model for tendon basic research and even for scaffold-free T/L engineering.


Subject(s)
Cell Culture Techniques , Fibroblasts , Ligaments , Organoids , Tendons , Humans , Tendons/cytology , Fibroblasts/cytology , Organoids/cytology , Ligaments/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Dermis/cytology
20.
Bone ; 187: 117197, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38986825

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) are integral to bone remodeling and homeostasis, as they are capable of differentiating into osteogenic and adipogenic lineages. This differentiation is substantially influenced by mechanosensitivity, particularly to tensile strain, which is a prevalent mechanical stimulus known to enhance osteogenic differentiation. This review specifically examines the effects of various cyclic tensile stress (CTS) conditions on BMSC osteogenesis. It delves into the effects of different loading devices, magnitudes, frequencies, elongation levels, dimensionalities, and coculture conditions, providing a comparative analysis that aids identification of the most conducive parameters for the osteogenic differentiation of BMSCs. Subsequently, this review delineates the signaling pathways activated by CTS, such as Wnt/ß-catenin, BMP, Notch, MAPK, PI3K/Akt, and Hedgehog, which are instrumental in mediating the osteogenic differentiation of BMSCs. Through a detailed examination of these pathways, this study elucidates the intricate mechanisms whereby tensile strain promotes osteogenic differentiation, offering valuable guidance for optimizing therapeutic strategies aimed at enhancing bone regeneration.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Cell Differentiation/physiology , Humans , Animals , Tensile Strength , Stress, Mechanical , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL