Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32.926
Filtrar
3.
J Virol ; 98(3): e0180223, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38334329

RESUMO

With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic. IMPORTANCE: Utilizing a human iPSC-derived kidney organoid model with improved proximal tubule (PT) maturity, we identified the mechanism of SARS-CoV-2 entry in renal cells, confirming ACE2 as the sole receptor and revealing redundancy in downstream cell surface TMPRSS- and endocytic Cathepsin-mediated pathways. In addition, these data address the implications of SARS-CoV-2 exposure in the setting of the commonly prescribed ACE-inhibitor, lisinopril, confirming its negligible impact on infection of kidney cells. Taken together, these results provide valuable insight into the mechanism of viral infection in the human kidney.


Assuntos
Enzima de Conversão de Angiotensina 2 , Rim , Organoides , SARS-CoV-2 , Internalização do Vírus , Humanos , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/complicações , COVID-19/virologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/virologia , Lisinopril/farmacologia , Lisinopril/metabolismo , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Organoides/virologia , Pandemias , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus/efeitos dos fármacos , Peptidil Dipeptidase A/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/virologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/virologia , Receptores de Coronavírus/metabolismo , Modelos Biológicos , Serina Endopeptidases/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Endossomos/virologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco/citologia
4.
Nucleic Acids Res ; 52(5): 2306-2322, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38142439

RESUMO

Spermatogonial stem cells functionality reside in the slow-cycling and heterogeneous undifferentiated spermatogonia cell population. This pool of cells supports lifelong fertility in adult males by balancing self-renewal and differentiation to produce haploid gametes. However, the molecular mechanisms underpinning long-term stemness of undifferentiated spermatogonia during adulthood remain unclear. Here, we discover that an epigenetic regulator, Polycomb repressive complex 1 (PRC1), shields adult undifferentiated spermatogonia from differentiation, maintains slow cycling, and directs commitment to differentiation during steady-state spermatogenesis in adults. We show that PRC2-mediated H3K27me3 is an epigenetic hallmark of adult undifferentiated spermatogonia. Indeed, spermatogonial differentiation is accompanied by a global loss of H3K27me3. Disruption of PRC1 impairs global H3K27me3 deposition, leading to precocious spermatogonial differentiation. Therefore, PRC1 directs PRC2-H3K27me3 deposition to maintain the self-renewing state of undifferentiated spermatogonia. Importantly, in contrast to its role in other tissue stem cells, PRC1 negatively regulates the cell cycle to maintain slow cycling of undifferentiated spermatogonia. Our findings have implications for how epigenetic regulators can be tuned to regulate the stem cell potential, cell cycle and differentiation to ensure lifelong fertility in adult males.


Assuntos
Complexo Repressor Polycomb 1 , Complexo Repressor Polycomb 2 , Espermatogênese , Células-Tronco , Humanos , Masculino , Diferenciação Celular , Histonas/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Espermatogônias , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Camundongos , Feminino , Complexo Repressor Polycomb 2/metabolismo
5.
Stem Cell Res Ther ; 14(1): 336, 2023 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-37981699

RESUMO

BACKGROUND: Kidney organoids derived from human pluripotent stem cells (HiPSCs) hold huge applications for drug screening, disease modeling, and cell transplanting therapy. However, these applications are limited since kidney organoid cannot maintain complete morphology and function like human kidney. Kidney organoids are not well differentiated since the core of the organoid lacked oxygen, nutrition, and vasculature, which creates essential niches. Hypoxia-inducible factor-1 α (HIF-1α) serves as a critical regulator in vascularization and cell survival under hypoxia environment. Less is known about the role of HIF-1α in kidney organoids in this regard. This study tried to investigate the effect of HIF-1α in kidney organoid vascularization and related disease modeling. METHODS: For the vascularization study, kidney organoids were generated from human induced pluripotent stem cells. We overexpressed HIF-1α via plasmid transfection or treated DMOG (Dimethyloxallyl Glycine, an agent for HIF-1α stabilization and accumulation) in kidney progenitor cells to detect the endothelium. For the disease modeling study, we treated kidney organoid with cisplatin under hypoxia environment, with additional HIF-1α transfection. RESULT: HIF-1α overexpression elicited kidney organoid vascularization. The endothelial cells and angiotool analysis parameters were increased in HIF-1α plasmid-transfected and DMOG-treated organoids. These angiogenesis processes were partially blocked by VEGFR inhibitors, semaxanib or axitinib. Cisplatin-induced kidney injury (Cleaved caspase 3) was protected by HIF-1α through the upregulation of CD31 and SOD2. CONCLUSION: We demonstrated that HIF-1α elicited the process of kidney organoid vascularization and protected against cisplatin-induced kidney organoid injury in hypoxia environment.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Rim , Modelos Biológicos , Organoides , Organoides/irrigação sanguínea , Organoides/metabolismo , Rim/metabolismo , Células-Tronco Multipotentes , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Humanos , Plasmídeos/genética , Expressão Gênica , Células-Tronco/citologia , Células-Tronco/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , /fisiopatologia , Axitinibe/farmacologia , Células Cultivadas , Cisplatino/farmacologia , Hipóxia Celular , Nefropatias/fisiopatologia
6.
Commun Biol ; 6(1): 1121, 2023 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-37925530

RESUMO

Skeletal muscle stem cells (MuSC) are crucial for tissue homoeostasis and repair after injury. Following activation, they proliferate to generate differentiating myoblasts. A proportion of cells self-renew, re-enter the MuSC niche under the basal lamina outside the myofiber and become quiescent. Quiescent MuSC have a primary cilium, which is disassembled upon cell cycle entry. Ex vivo experiments suggest cilia are important for MuSC self-renewal, however, their requirement for muscle regeneration in vivo remains poorly understood. Talpid3 (TA3) is essential for primary cilia formation and Hedgehog (Hh) signalling. Here we use tamoxifen-inducible conditional deletion of TA3 in MuSC (iSC-KO) and show that regeneration is impaired in response to cytotoxic injury. Depletion of MuSC after regeneration suggests impaired self-renewal, also consistent with an exacerbated phenotype in TA3iSC-KO mice after repeat injury. Single cell transcriptomics of MuSC progeny isolated from myofibers identifies components of several signalling pathways, which are deregulated in absence of TA3, including Hh and Wnt. Pharmacological activation of Wnt restores muscle regeneration, while purmorphamine, an activator of the Smoothened (Smo) co-receptor in the Hh pathway, has no effect. Together, our data show that TA3 and primary cilia are important for MuSC self-renewal and pharmacological treatment can efficiently restore muscle regeneration.


Assuntos
Proteínas de Ciclo Celular , Cílios , Músculos , Células Satélites de Músculo Esquelético , Células-Tronco , Animais , Camundongos , Células Cultivadas , Cílios/genética , Cílios/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Músculos/citologia , Células Satélites de Músculo Esquelético/metabolismo , Proteínas de Ciclo Celular/genética , Células-Tronco/citologia
7.
J Cell Biol ; 222(12)2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-37843535

RESUMO

With each breath, oxygen diffuses across remarkably thin alveolar type I (AT1) cells into underlying capillaries. Interspersed cuboidal AT2 cells produce surfactant and act as stem cells. Even transient disruption of this delicate barrier can promote capillary leak. Here, we selectively ablated AT1 cells, which uncovered rapid AT2 cell flattening with near-continuous barrier preservation, culminating in AT1 differentiation. Proliferation subsequently restored depleted AT2 cells in two phases, mitosis of binucleated AT2 cells followed by replication of mononucleated AT2 cells. M phase entry of binucleated and S phase entry of mononucleated cells were both triggered by AT1-produced hbEGF signaling via EGFR to Wnt-active AT2 cells. Repeated AT1 cell killing elicited exuberant AT2 proliferation, generating aberrant daughter cells that ceased surfactant function yet failed to achieve AT1 differentiation. This hyperplasia eventually resolved, yielding normal-appearing alveoli. Overall, this specialized regenerative program confers a delicate simple epithelium with functional resiliency on par with the physical durability of thicker, pseudostratified, or stratified epithelia.


Assuntos
Pulmão , Células-Tronco , Tensoativos , Diferenciação Celular , Divisão Celular , Células Cultivadas , Células-Tronco/citologia
9.
Prog Mol Biol Transl Sci ; 199: 109-127, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37678968

RESUMO

Conventional two-dimensional (2-D) cultivation are easy to utilize for human pluripotent stem (hPS) cell cultivation in standard techniques and are important for analysis or development of the signal pathways to keep pluripotent state of hPS cells cultivated on 2-D cell culture materials. However, the most efficient protocol to prepare hPS cells is the cell culture in a three dimensional (3-D) cultivation unit because huge numbers of hPS cells should be utilized in clinical treatment. Some 3-D cultivation strategies for hPS cells are considered: (a) microencapsulated cell cultivation in suspended hydrogels, (b) cell cultivation on microcarriers (MCs), (c) cell cultivation on self-aggregated spheroid [cell aggregates; embryoid bodies (EBs) and organoids], (d) cell cultivation on microfibers or nanofibers, and (e) cell cultivation in macroporous scaffolds. These cultivation ways are described in this chapter.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Diferenciação Celular , Células-Tronco , Humanos , Células-Tronco/citologia , Técnicas de Cultura de Células , Técnicas de Cultura de Células em Três Dimensões/métodos , Hepatócitos/citologia , Hidrogéis , Encapsulamento de Células , Tecidos Suporte , Engenharia Tecidual
10.
Nature ; 621(7979): 602-609, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37704733

RESUMO

Vertebral bone is subject to a distinct set of disease processes from long bones, including a much higher rate of solid tumour metastases1-4. The basis for this distinct biology of vertebral bone has so far remained unknown. Here we identify a vertebral skeletal stem cell (vSSC) that co-expresses ZIC1 and PAX1 together with additional cell surface markers. vSSCs display formal evidence of stemness, including self-renewal, label retention and sitting at the apex of their differentiation hierarchy. vSSCs are physiologic mediators of vertebral bone formation, as genetic blockade of the ability of vSSCs to generate osteoblasts results in defects in the vertebral neural arch and body. Human counterparts of vSSCs can be identified in vertebral endplate specimens and display a conserved differentiation hierarchy and stemness features. Multiple lines of evidence indicate that vSSCs contribute to the high rates of vertebral metastatic tropism observed in breast cancer, owing in part to increased secretion of the novel metastatic trophic factor MFGE8. Together, our results indicate that vSSCs are distinct from other skeletal stem cells and mediate the unique physiology and pathology of vertebrae, including contributing to the high rate of vertebral metastasis.


Assuntos
Neoplasias da Mama , Linhagem da Célula , Metástase Neoplásica , Coluna Vertebral , Células-Tronco , Humanos , Neoplasias da Mama/patologia , Diferenciação Celular , Autorrenovação Celular , Metástase Neoplásica/patologia , Osteoblastos/citologia , Osteoblastos/patologia , Coluna Vertebral/citologia , Coluna Vertebral/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Biomarcadores
11.
BMC Biol ; 21(1): 169, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553612

RESUMO

BACKGROUND: The nutrient-absorbing villi of small intestines are renewed and repaired by intestinal stem cells (ISCs), which reside in a well-organized crypt structure. Genetic studies have shown that Wnt molecules secreted by telocytes, Gli1+ stromal cells, and epithelial cells are required for ISC proliferation and villus homeostasis. Intestinal stromal cells are heterogeneous and single-cell profiling has divided them into telocytes/subepithelial myofibroblasts, myocytes, pericytes, trophocytes, and Pdgfralow stromal cells. Yet, the niche function of these stromal populations remains incompletely understood. RESULTS: We show here that a Twist2 stromal lineage, which constitutes the Pdgfralow stromal cell and trophocyte subpopulations, maintains the crypt structure to provide an inflammation-restricting niche for regenerating ISCs. Ablating Twist2 lineage cells or deletion of one Wntless allele in these cells disturbs the crypt structure and impairs villus homeostasis. Upon radiation, Wntless haplo-deficiency caused decreased production of anti-microbial peptides and increased inflammation, leading to defective ISC proliferation and crypt regeneration, which were partially rescued by eradication of commensal bacteria. In addition, we show that Wnts secreted by Acta2+ subpopulations also play a role in crypt regeneration but not homeostasis. CONCLUSIONS: These findings suggest that ISCs may require different niches for villus homeostasis and regeneration and that the Twist2 lineage cells may help to maintain a microbe-restricted environment to allow ISC-mediated crypt regeneration.


Assuntos
Linhagem da Célula , Homeostase , Intestinos , Nicho de Células-Tronco , Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Intestinos/citologia , Intestinos/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Animais , Camundongos
12.
Nature ; 620(7972): 145-153, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37468639

RESUMO

Human-specific genomic changes contribute to the unique functionalities of the human brain1-5. The cellular heterogeneity of the human brain6,7 and the complex regulation of gene expression highlight the need to characterize human-specific molecular features at cellular resolution. Here we analysed single-nucleus RNA-sequencing and single-nucleus assay for transposase-accessible chromatin with sequencing datasets for human, chimpanzee and rhesus macaque brain tissue from posterior cingulate cortex. We show a human-specific increase of oligodendrocyte progenitor cells and a decrease of mature oligodendrocytes across cortical tissues. Human-specific regulatory changes were accelerated in oligodendrocyte progenitor cells, and we highlight key biological pathways that may be associated with the proportional changes. We also identify human-specific regulatory changes in neuronal subtypes, which reveal human-specific upregulation of FOXP2 in only two of the neuronal subtypes. We additionally identify hundreds of new human accelerated genomic regions associated with human-specific chromatin accessibility changes. Our data also reveal that FOS::JUN and FOX motifs are enriched in the human-specifically accessible chromatin regions of excitatory neuronal subtypes. Together, our results reveal several new mechanisms underlying the evolutionary innovation of human brain at cell-type resolution.


Assuntos
Evolução Molecular , Giro do Cíngulo , Animais , Humanos , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Conjuntos de Dados como Assunto , Genoma Humano/genética , Genômica , Giro do Cíngulo/citologia , Giro do Cíngulo/metabolismo , Macaca mulatta/genética , Neurônios/classificação , Neurônios/citologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Pan troglodytes/genética , Análise da Expressão Gênica de Célula Única , Células-Tronco/citologia , Transposases/metabolismo , Montagem e Desmontagem da Cromatina
13.
BMC Oral Health ; 23(1): 501, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37468947

RESUMO

BACKGROUND: The key to the success of endogenous regeneration is to improve the homing rate of stem cells, and low-energy laser is an effective auxiliary means to promote cell migration and proliferation. The purpose of this study was to observe whether low-energy neodymium (Nd: YAG) laser with appropriate parameters can affect the proliferation and migration of periodontal ligament stem cells (PDLSCs) through SDF-1/CXCR4 pathway. METHODS: h PDLSCs were cultured and identified. CCK8 assay was used to detect the proliferation of h PDLSCs after different power (0, 0.25, 0.5, 1, and 1.5 W) Nd: YAG laser (MSP, 10 Hz, 30 s, 300 µ m) irradiation at 2th, 3rd,5th, and 7th days, and the optimal laser irradiation parameters were selected for subsequent experiments. Then, the cells were categorized into five groups: control group (C), SDF-1 group (S), AMD3100 group (A), Nd: YAG laser irradiation group (N), and Nd: YAG laser irradiation + AMD3100 group (N + A). the migration of h PDLSCs was observed using Transwell, and the SDF-1 expression was evaluated using ELISA andRT-PCR. The SPSS Statistics 21.0 software was used for statistical analysis. RESULTS: The fibroblasts cultured were identified as h PDLSCs. Compared with the C, when the power was 1 W, the proliferation rate of h PDLSCs was accelerated (P < 0.05). When the power was 1.5 W, the proliferation rate decreased (P < 0.05). When the power was 0.25 and 0.5 W, no statistically significant difference in the proliferation rate was observed (P > 0.05). The number of cell perforations values as follows: C (956.5 ± 51.74), A (981.5 ± 21.15), S (1253 ± 87.21), N (1336 ± 48.54), and N + A (1044 ± 22.13), that increased significantly in group N (P < 0.05), but decreased in group N + A (P < 0.05). The level of SDF-1 and the expression level of SDF-1 mRNA in groups N and N + A was higher than that in group C (P < 0.05) but lower than that in group A (P < 0.05). CONCLUSIONS: Nd: YAG laser irradiation with appropriate parameters provides a new method for endogenous regeneration of periodontal tissue. SDF-1/CXCR4 signaling pathway may be the mechanism of LLLT promoting periodontal regeneration.


Assuntos
Movimento Celular , Lasers de Estado Sólido , Ligamento Periodontal , Células-Tronco , Humanos , Benzilaminas , Ligamento Periodontal/citologia , Receptores CXCR4 , Células-Tronco/citologia
14.
ACS Appl Mater Interfaces ; 15(27): 32148-32161, 2023 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-37364369

RESUMO

Synthetic matrices that are cytocompatible, cell adhesive, and cell responsive are needed for the engineering of implantable, secretory salivary gland constructs to treat radiation induced xerostomia or dry mouth. Here, taking advantage of the bioorthogonality of the Michael-type addition reaction, hydrogels with comparable stiffness but varying degrees of degradability (100% degradable, 100DEG; 50% degradable, 50DEG; and nondegradable, 0DEG) by cell-secreted matrix metalloproteases (MMPs) were synthesized using thiolated HA (HA-SH), maleimide (MI)-conjugated integrin-binding peptide (RGD-MI), and MI-functionalized peptide cross-linkers that are protease degradable (GIW-bisMI) or nondegradable (GIQ-bisMI). Organized multicellular structures developed readily in all hydrogels from dispersed primary human salivary gland stem cells (hS/PCs). As the matrix became progressively degradable, cells proliferated more readily, and the multicellular structures became larger, less spherical, and more lobular. Immunocytochemical analysis showed positive staining for stem/progenitor cell markers CD44 and keratin 5 (K5) in all three types of cultures and positive staining for the acinar marker α-amylase under 50DEG and 100DEG conditions. Quantitatively at the mRNA level, the expression levels of key stem/progenitor markers KIT, KRT5, and ETV4/5 were significantly increased in the degradable gels as compared to the nondegradable counterparts. Western blot analyses revealed that imparting matrix degradation led to >3.8-fold increase in KIT expression by day 15. The MMP-degradable hydrogels also promoted the development of a secretary phenotype, as evidenced by the upregulation of acinar markers α-amylase (AMY), aquaporin-5 (AQP5), and sodium-potassium chloride cotransporter 1 (SLC12A2). Collectively, we show that cell-mediated matrix remodeling is necessary for the development of regenerative pro-acinar progenitor cells from hS/PCs.


Assuntos
Glândulas Salivares , Células-Tronco , Humanos , Células Cultivadas , Glândulas Salivares/citologia , Glândulas Salivares/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Hidrogéis/química , Compostos de Sulfidrila/química , Sobrevivência Celular , Biomarcadores
15.
Nature ; 618(7966): 808-817, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37344645

RESUMO

Niche signals maintain stem cells in a prolonged quiescence or transiently activate them for proper regeneration1. Altering balanced niche signalling can lead to regenerative disorders. Melanocytic skin nevi in human often display excessive hair growth, suggesting hair stem cell hyperactivity. Here, using genetic mouse models of nevi2,3, we show that dermal clusters of senescent melanocytes drive epithelial hair stem cells to exit quiescence and change their transcriptome and composition, potently enhancing hair renewal. Nevus melanocytes activate a distinct secretome, enriched for signalling factors. Osteopontin, the leading nevus signalling factor, is both necessary and sufficient to induce hair growth. Injection of osteopontin or its genetic overexpression is sufficient to induce robust hair growth in mice, whereas germline and conditional deletions of either osteopontin or CD44, its cognate receptor on epithelial hair cells, rescue enhanced hair growth induced by dermal nevus melanocytes. Osteopontin is overexpressed in human hairy nevi, and it stimulates new growth of human hair follicles. Although broad accumulation of senescent cells, such as upon ageing or genotoxic stress, is detrimental for the regenerative capacity of tissue4, we show that signalling by senescent cell clusters can potently enhance the activity of adjacent intact stem cells and stimulate tissue renewal. This finding identifies senescent cells and their secretome as an attractive therapeutic target in regenerative disorders.


Assuntos
Cabelo , Melanócitos , Transdução de Sinais , Animais , Camundongos , Cabelo/citologia , Cabelo/crescimento & desenvolvimento , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Receptores de Hialuronatos/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Nevo/metabolismo , Nevo/patologia , Osteopontina/metabolismo , Células-Tronco/citologia
16.
J Virol ; 97(6): e0068923, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37289083

RESUMO

Goblet cells and their secreted mucus are important elements of the intestinal mucosal barrier, which allows host cells to resist invasion by intestinal pathogens. Porcine deltacoronavirus (PDCoV) is an emerging swine enteric virus that causes severe diarrhea in pigs and causes large economic losses to pork producers worldwide. To date, the molecular mechanisms by which PDCoV regulates the function and differentiation of goblet cells and disrupts the intestinal mucosal barrier remain to be determined. Here, we report that in newborn piglets, PDCoV infection disrupts the intestinal barrier: specifically, there is intestinal villus atrophy, crypt depth increases, and tight junctions are disrupted. There is also a significant reduction in the number of goblet cells and the expression of MUC-2. In vitro, using intestinal monolayer organoids, we found that PDCoV infection activates the Notch signaling pathway, resulting in upregulated expression of HES-1 and downregulated expression of ATOH-1 and thereby inhibiting the differentiation of intestinal stem cells into goblet cells. Our study shows that PDCoV infection activates the Notch signaling pathway to inhibit the differentiation of goblet cells and their mucus secretion, resulting in disruption of the intestinal mucosal barrier. IMPORTANCE The intestinal mucosal barrier, mainly secreted by the intestinal goblet cells, is a crucial first line of defense against pathogenic microorganisms. PDCoV regulates the function and differentiation of goblet cells, thereby disrupting the mucosal barrier; however, the mechanism by which PDCoV disrupts the barrier is not known. Here, we report that in vivo, PDCoV infection decreases villus length, increases crypt depth, and disrupts tight junctions. Moreover, PDCoV activates the Notch signaling pathway, inhibiting goblet cell differentiation and mucus secretion in vivo and in vitro. Thus, our results provide a novel insight into the mechanism underlying intestinal mucosal barrier dysfunction caused by coronavirus infection.


Assuntos
Infecções por Coronavirus , Células Caliciformes , Receptores Notch , Doenças dos Suínos , Animais , Coronavirus , Infecções por Coronavirus/patologia , Infecções por Coronavirus/veterinária , Células Caliciformes/citologia , Transdução de Sinais , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Células-Tronco/citologia , Diferenciação Celular , Receptores Notch/metabolismo
17.
Acta Biomater ; 166: 109-118, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37245640

RESUMO

Skeletal progenitor: collagen interactions are critical for bone development and regeneration. Both collagen-binding integrins and discoidin domain receptors (DDR1 and DDR2) function as collagen receptors in bone. Each receptor is activated by a distinct collagen sequence; GFOGER for integrins and GVMGFO for DDRs. Specific triple helical peptides containing each of these binding domains were evaluated for ability to stimulate DDR2 and integrin signaling and osteoblast differentiation. GVMGFO peptide stimulated DDR2 Y740 phosphorylation and osteoblast differentiation as measured by induction of osteoblast marker mRNAs and mineralization without affecting integrin activity. In contrast, GFOGER peptide stimulated focal adhesion kinase (FAK) Y397 phosphorylation, an early measure of integrin activation, and to a lesser extent osteoblast differentiation without affecting DDR2-P. Significantly, the combination of both peptides cooperatively enhanced both DDR2 and FAK signaling and osteoblast differentiation, a response that was blocked in Ddr2-deficient cells. These studies suggest that the development of scaffolds containing DDR and integrin-activating peptides may provide a new route for promoting bone regeneration. STATEMENT OF SIGNIFICANCE: A method for stimulating osteoblast differentiation of skeletal progenitor cells is described that uses culture surfaces coated with a collagen-derived triple-helical peptide to selectively activate discoidin domain receptors. When this peptide is combined with an integrin-activating peptide, synergistic stimulation of differentiation is seen. This approach of combining collagen-derived peptides to stimulate the two main collagen receptors in bone (DDR2 and collagen-binding integrins) provides a route for developing a new class of tissue engineering scaffolds for bone regeneration.


Assuntos
Diferenciação Celular , Osteoblastos , Células-Tronco , Animais , Camundongos , Linhagem Celular , Colágeno/química , Peptídeos/química , Peptídeos/farmacologia , Receptor com Domínio Discoidina 2/química , Integrinas/química , Células-Tronco/citologia , Osteoblastos/citologia , Diferenciação Celular/efeitos dos fármacos , Fosforilação , Transdução de Sinais/efeitos dos fármacos
18.
Adv Biol (Weinh) ; 7(8): e2200332, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37236203

RESUMO

Hydrogels are 3D scaffolds used as alternatives to in vivo models for disease modeling and delivery of cells and drugs. Existing hydrogel classifications include synthetic, recombinant, chemically defined, plant- or animal-based, and tissue-derived matrices. There is a need for materials that can support both human tissue modeling and clinically relevant applications requiring stiffness tunability. Human-derived hydrogels are not only clinically relevant, but they also minimize the use of animal models for pre-clinical studies. This study aims to characterize XGel, a new human-derived hydrogel as an alternative to current murine-derived and synthetic recombinant hydrogels that features unique physiochemical, biochemical, and biological properties that support adipocyte and bone differentiation. Rheology studies determine the viscosity, stiffness, and gelation features of XGel. Quantitative studies for quality control support consistency in the protein content between lots. Proteomics studies reveal that XGel is predominantly composed of extracellular matrix proteins, including fibrillin, collagens I-VI, and fibronectin. Electron microscopy of the hydrogel provides phenotypic characteristics in terms of porosity and fiber size. The hydrogel demonstrates biocompatibility as a coating material and as a 3D scaffold for the growth of multiple cell types. The results provide insight into the biological compatibility of this human-derived hydrogel for tissue engineering.


Assuntos
Hidrogéis , Células-Tronco , Engenharia Tecidual , Hidrogéis/química , Humanos , Matriz Extracelular , Proliferação de Células , Células-Tronco/citologia
20.
Nature ; 616(7958): 774-782, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37076619

RESUMO

For unknow reasons, the melanocyte stem cell (McSC) system fails earlier than other adult stem cell populations1, which leads to hair greying in most humans and mice2,3. Current dogma states that McSCs are reserved in an undifferentiated state in the hair follicle niche, physically segregated from differentiated progeny that migrate away following cues of regenerative stimuli4-8. Here we show that most McSCs toggle between transit-amplifying and stem cell states for both self-renewal and generation of mature progeny, a mechanism fundamentally distinct from those of other self-renewing systems. Live imaging and single-cell RNA sequencing revealed that McSCs are mobile, translocating between hair follicle stem cell and transit-amplifying compartments where they reversibly enter distinct differentiation states governed by local microenvironmental cues (for example, WNT). Long-term lineage tracing demonstrated that the McSC system is maintained by reverted McSCs rather than by reserved stem cells inherently exempt from reversible changes. During ageing, there is accumulation of stranded McSCs that do not contribute to the regeneration of melanocyte progeny. These results identify a new model whereby dedifferentiation is integral to homeostatic stem cell maintenance and suggest that modulating McSC mobility may represent a new approach for the prevention of hair greying.


Assuntos
Desdiferenciação Celular , Folículo Piloso , Melanócitos , Nicho de Células-Tronco , Células-Tronco , Animais , Humanos , Camundongos , Folículo Piloso/citologia , Melanócitos/citologia , Células-Tronco/citologia , Microambiente Celular , Linhagem da Célula , Envelhecimento , Homeostase , Cor de Cabelo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...