Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38814743

ABSTRACT

Apical expansion of calvarial osteoblast progenitors from the cranial mesenchyme (CM) above the eye is integral to calvarial growth and enclosure of the brain. The cellular behaviors and signals underlying the morphogenetic process of calvarial expansion are unknown. Time-lapse light-sheet imaging of mouse embryos revealed calvarial progenitors intercalate in 3D in the CM above the eye, and exhibit protrusive and crawling activity more apically. CM cells express non-canonical Wnt/planar cell polarity (PCP) core components and calvarial osteoblasts are bidirectionally polarized. We found non-canonical ligand Wnt5a-/- mutants have less dynamic cell rearrangements and protrusive activity. Loss of CM-restricted Wntless (CM-Wls), a gene required for secretion of all Wnt ligands, led to diminished apical expansion of Osx+ calvarial osteoblasts in the frontal bone primordia in a non-cell autonomous manner without perturbing proliferation or survival. Calvarial osteoblast polarization, progressive cell elongation and enrichment for actin along the baso-apical axis were dependent on CM-Wnts. Thus, CM-Wnts regulate cellular behaviors during calvarial morphogenesis for efficient apical expansion of calvarial osteoblasts. These findings also offer potential insights into the etiologies of calvarial dysplasias.


Subject(s)
Mesoderm , Morphogenesis , Osteoblasts , Skull , Wnt Proteins , Animals , Osteoblasts/metabolism , Osteoblasts/cytology , Skull/embryology , Mice , Mesoderm/cytology , Mesoderm/metabolism , Wnt Proteins/metabolism , Wnt Proteins/genetics , Cell Polarity , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Cell Movement , Cell Proliferation
2.
Proteomics ; : e2300641, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38629187

ABSTRACT

Human embryonic stem cells (hESCs) resemble the pluripotent epiblast cells found in the early postimplantation human embryo and represent the "primed" state of pluripotency. One factor that helps primed pluripotent cells retain pluripotency and prepare genes for differentiation is the transcription factor TCF7L1, a member of a small family of proteins known as T cell factors/Lymphoid enhancer factors (TCF/LEF) that act as downstream components of the WNT signaling pathway. Transcriptional output of the WNT pathway is regulated, in part, by the activity of TCF/LEFs in conjunction with another component of the WNT pathway, ß-CATENIN. Because TCF7L1 plays an important role in regulating pluripotency, we began to characterize the protein complex associated with TCF7L1 when bound to chromatin in hESCs using rapid immunoprecipitation of endogenous proteins (RIME).  Data are available via ProteomeXchange with identifier PXD047582. These data identify known and new partners of TCF7L1 on chromatin and provide novel insights into how TCF7L1 and pluripotency itself might be regulated.

3.
Elife ; 122023 05 16.
Article in English | MEDLINE | ID: mdl-37191285

ABSTRACT

Fenestrated and blood-brain barrier (BBB)-forming endothelial cells constitute major brain capillaries, and this vascular heterogeneity is crucial for region-specific neural function and brain homeostasis. How these capillary types emerge in a brain region-specific manner and subsequently establish intra-brain vascular heterogeneity remains unclear. Here, we performed a comparative analysis of vascularization across the zebrafish choroid plexuses (CPs), circumventricular organs (CVOs), and retinal choroid, and show common angiogenic mechanisms critical for fenestrated brain capillary formation. We found that zebrafish deficient for Gpr124, Reck, or Wnt7aa exhibit severely impaired BBB angiogenesis without any apparent defect in fenestrated capillary formation in the CPs, CVOs, and retinal choroid. Conversely, genetic loss of various Vegf combinations caused significant disruptions in Wnt7/Gpr124/Reck signaling-independent vascularization of these organs. The phenotypic variation and specificity revealed heterogeneous endothelial requirements for Vegfs-dependent angiogenesis during CP and CVO vascularization, identifying unexpected interplay of Vegfc/d and Vegfa in this process. Mechanistically, expression analysis and paracrine activity-deficient vegfc mutant characterization suggest that endothelial cells and non-neuronal specialized cell types present in the CPs and CVOs are major sources of Vegfs responsible for regionally restricted angiogenic interplay. Thus, brain region-specific presentations and interplay of Vegfc/d and Vegfa control emergence of fenestrated capillaries, providing insight into the mechanisms driving intra-brain vascular heterogeneity and fenestrated vessel formation in other organs.


Subject(s)
Endothelial Cells , Zebrafish , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Capillaries , Endothelial Cells/physiology , Neovascularization, Physiologic/genetics , Zebrafish/genetics
4.
Curr Top Dev Biol ; 150: 91-128, 2022.
Article in English | MEDLINE | ID: mdl-35817507

ABSTRACT

Wnts are a family of secreted, lipid-modified signaling glycoproteins that regulate a multiplicity of fundamental biological processes. Wnt signaling is essential for embryonic development, controlling body axis patterning, cell proliferation, cell migration and cell fate specification needed for proper tissue and organ formation. In adulthood, Wnt signaling controls tissue regeneration in a wide range of organs, and disturbance of this system is common in cancer and other diseases. A key feature of Wnt signaling is that it is a local process. Wnts signal via paracrine, cell-to-cell communication. Upon synthesis and transport to the plasma membrane in the "sending" cell, Wnts travel to nearby "receiving" cells. At the plasma membrane of these receiving cells, they interact with a variety of cell-surface receptors. This interaction triggers a diversity of different downstream signaling events, including the stabilization of ß-catenin and tissue-specific changes in gene expression. Wnt signaling is a local event because as an indispensable step in their maturation, Wnts are palmitoleated immediately after synthesis. This lipid modification is essential for Wnts to be transported and biologically active, but it also renders them highly hydrophobic. This makes all Wnts highly dependent on carrier proteins and specialized cellular structures both for intra- and inter-cellular movement. How this complex machinery acts in concert to deliver its highly important payload from the place of synthesis to the correct site of delivery is under active investigation. Here, we review the current understanding of how lipid-modified Wnts are processed, transported, and guided to their place of action.


Subject(s)
Wnt Proteins , Wnt Signaling Pathway , Body Patterning/genetics , Cell Movement , Lipids , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology
5.
Cell Stem Cell ; 29(7): 1067-1082.e18, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35728595

ABSTRACT

Barrier epithelia depend upon resident stem cells for homeostasis, defense, and repair. Epithelial stem cells of small and large intestines (ISCs) respond to their local microenvironments (niches) to fulfill a continuous demand for tissue turnover. The complexity of these niches and underlying communication pathways are not fully known. Here, we report a lymphatic network at the intestinal crypt base that intimately associates with ISCs. Employing in vivo loss of function and lymphatic:organoid cocultures, we show that crypt lymphatics maintain ISCs and inhibit their precocious differentiation. Pairing single-cell and spatial transcriptomics, we apply BayesPrism to deconvolve expression within spatial features and develop SpaceFold to robustly map the niche at high resolution, exposing lymphatics as a central signaling hub for the crypt in general and ISCs in particular. We identify WNT-signaling factors (WNT2, R-SPONDIN-3) and a hitherto unappreciated extracellular matrix protein, REELIN, as crypt lymphatic signals that directly govern the regenerative potential of ISCs.


Subject(s)
Intestines , Stem Cells , Cell Proliferation , Intestinal Mucosa/metabolism , Organoids , Signal Transduction , Wnt Proteins/metabolism
6.
Ecotoxicol Environ Saf ; 224: 112667, 2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34425536

ABSTRACT

Cadmium is a severe environmental pollutant that mainly targets kidney and causes kidney disease in the end. However, the mechanism of cadmium-induced kidney disease is still unclear. In this study, we treated SD rats, kidney epithelial or fibroblast cells with cadmium, and examined the renal fibrosis process and underlying cellular and molecular mechanism. Rats received daily (Monday-Friday) subcutaneous injections of CdCl2, 0.6 mg/kg, for 6 weeks or 12 weeks, and NRK-52E cells were treated with CdCl2 of 8 µM for 24 h. Sirius red staining and immunohistochemistry assay showed that sub-chronic exposure to cadmium caused interstitial fibrosis in rat kidneys. Cell experiments showed that cadmium treatment in NRK-52E cells only changed levels of α-SMA, vimentin and E-cadherin, but not collagen1, indicating that cells other than EMT cells might be responsible for the extracellular matrix production. By contrast, co-culture of NRK-49F cells with cadmium-treated NRK-52E cells produced collagen1. Assays of supernatant of NRK-52E cell culture showed that the secreted Wnt1, Wnt4 were increased, while miR-503-5p was decreased by cadmium treatment. RT-QPCR assay found that miR-503-5p was downregulated in both kidney of rats and NRK-52E cells exposed to cadmium. miR-503-5p was further shown to be competent in hindering epithelial-mesenchymal transition and fibroblast activation. Given the well established involvement of Wnt/ß-catenin pathway in fibrosis, this study suggested that dysregulations of Wnts and miR-503-5p coordinate in mediating cadmium-induced kidney fibrosis. Our findings might provide new insight in the cellular and molecular mechanisms of kidney interstitial fibrosis and novel therapeutic targets for cadmium-induced kidney disease.

7.
Front Endocrinol (Lausanne) ; 12: 667480, 2021.
Article in English | MEDLINE | ID: mdl-34108937

ABSTRACT

Wnt signaling plays a critical role in bone formation, homeostasis, and injury repair. Multiple cell types in bone have been proposed to produce the Wnts required for these processes. The specific role of Wnts produced from cells of hematopoietic origin has not been previously characterized. Here, we examined if hematopoietic Wnts play a role in physiological musculoskeletal development and in fracture healing. Wnt secretion from hematopoietic cells was blocked by genetic knockout of the essential Wnt modifying enzyme PORCN, achieved by crossing Vav-Cre transgenic mice with Porcnflox mice. Knockout mice were compared with their wild-type littermates for musculoskeletal development including bone quantity and quality at maturation. Fracture healing including callus quality and quantity was assessed in a diaphyseal fracture model using quantitative micro computer-assisted tomographic scans, histological analysis, as well as biomechanical torsional and 4-point bending stress tests. The hematopoietic Porcn knockout mice had normal musculoskeletal development, with normal bone quantity and quality on micro-CT scans of the vertebrae. They also had normal gross skeletal dimensions and normal bone strength. Hematopoietic Wnt depletion in the healing fracture resulted in fewer osteoclasts in the fracture callus, with a resultant delay in callus remodeling. All calluses eventually progressed to full maturation. Hematopoietic Wnts, while not essential, modulate osteoclast numbers during fracture healing. These osteoclasts participate in callus maturation and remodeling. This demonstrates the importance of diverse Wnt sources in bone repair.


Subject(s)
Acyltransferases/physiology , Bony Callus/cytology , Fracture Healing , Membrane Proteins/physiology , Osteoclasts/cytology , Osteogenesis , Wnt Signaling Pathway , Animals , Biomechanical Phenomena , Bony Callus/metabolism , Female , Male , Mice , Mice, Knockout , Osteoclasts/metabolism
8.
Mol Biol Rep ; 48(4): 3515-3527, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33881728

ABSTRACT

The role of copper and selenium on activation of estradiol synthesis pathways viz. PKA/AKT/WNT is not clearly elucidated. On this background we attempt to elcuiated the role of copper and selenium on mRNA expression of genes associated with estradiol synthesis in caprine ovarian granulose cell models. Ovarian granulosa cells from medium (3-5 mm) sized follicles were aspirated and distributed separately to different groups. Group I: control, Group II: cupric chloride (Cu: 0.5 mM), Group III: sodium selenite (Se: 100 ng/ml), Group IV: Cu + Se. The cells (105/well) were cultured in 96 well plate in the base culture medium of MEMα comprising of nonessential amino acids (1.1 mM), FSH (10 ng/mL), transferrin (5 µg/mL), IGF-I (2 ng/mL), androstenedione (10-6 M), penicillin (100 IU/mL), streptomycin (0.1 mg/mL) and fungizone (0.625 µl/mL) and insulin (1 ng/mL). The cells were incubated in a carbondioxide incubator (38 °C, 5% CO2, 95% RH). The medium was changed on alternate days and cells were harvested on day 6. Day 6 media was used for estimation of estradiol. The RNA isolated form harvested cells was used for qPCR assay. There was no significant (p > 0.05) difference in estradiol concentration between groups. The mRNA expression of AKT1, CYP19A1, WNT2 & 4, FZD6 and APC2 were significantly (p < 0.05) higher in Cu and Cu + Se groups compared to control. Whereas, the mRNA transcript of DVL1 and CSNK1 was significantly (p < 0.05) higher in Cu + Se group compared to control. Incontrast, no significant difference in mRNA expression of PRKAR1A and CTNNB1 was noticed. Our study support a key role of copper and selenium in activation of AKT and WNT signalling pathway that further lead to increase in the mRNA expression of CYP19A1.


Subject(s)
Aromatase/genetics , Copper/pharmacology , Granulosa Cells/metabolism , Selenium/pharmacology , Animals , Aromatase/metabolism , Cells, Cultured , Female , Goats , Granulosa Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wnt Signaling Pathway
9.
J Cell Mol Med ; 25(4): 1958-1971, 2021 02.
Article in English | MEDLINE | ID: mdl-33434361

ABSTRACT

The activation of Wnt/ß-catenin pathway plays a pivotal role in promoting renal fibrosis. The activation of Wnt/ß-catenin pathway relies on the binding of Wnts to Frizzled receptors on cell membrane. However, the factor regulating Wnts production remains unclear. Here, we demonstrated that transcriptional factor FoxM1 was significantly increased in obstructed kidneys and patients' kidneys with fibrosis. The up-regulation of FoxM1 mainly distributed in tubular epithelial cells. Pharmacological inhibition of FoxM1 down-regulated multi-Wnts elevation in UUO mice and attenuated renal fibrosis. In cultured renal tubular epithelial cells, overexpression of FoxM1 promoted 8 Wnts expression, while knock-down on FoxM1-suppressed multi-Wnts including Wnt1, Wnt2b and Wnt3 expression induced by Ang II. Chromatin immunoprecipitation PCR confirmed that FoxM1 bound to Wnt1, Wnt2b, Wnt3 promoters and luciferase assay further identified that the transcriptions of Wnt1, Wnt2b and Wnt3 were regulated by FoxM1. Thus, our findings show that multi-Wnt family members were regulated by transcriptional factor FoxM1. FoxM1 might be a key switch for activating ß-catenin pathway and renal fibrosis. Therefore, FoxM1 might be a potential therapeutic target in manipulating renal fibrosis.


Subject(s)
Forkhead Box Protein M1/metabolism , Gene Expression Regulation , Kidney Diseases/genetics , Kidney Diseases/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Biomarkers , Cells, Cultured , Disease Models, Animal , Disease Susceptibility , Epithelial Cells/metabolism , Fibrosis , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/genetics , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Kidney Diseases/pathology , Kidney Tubules/metabolism , Male , Mice
10.
Dev Dyn ; 250(3): 360-376, 2021 03.
Article in English | MEDLINE | ID: mdl-32738003

ABSTRACT

More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/metabolism , Chondrogenesis , Osteoarthritis , Regeneration , Animals , Humans , Osteoarthritis/metabolism , Osteoarthritis/physiopathology
11.
Neurosci Lett ; 742: 135520, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33246026

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of midbrain dopaminergic (DA) neurons. Neural stem cells (NSCs) are the most promising cells for cell-replacement therapy for PD. However, the poor differentiation and maturation of DA neurons and decreased cell survival after transplantation are a challenge. Tetrahydroxystilbene glucoside (2,3,5,4'-tetrahydroxystilbene-2-O-glucoside; TSG), an active component of the popular traditional Chinese medicinal plant Polygonum multiflorum Thunb, possesses multiple pharmacological actions. In this study, we determined whether TSG can induce neural stem cell (NSCs) differentiation into neurons, especially DA neurons, and the possible involvement of Wnt/ß-catenin signaling pathways. Results revealed that NSCs differentiated primarily into astrocytes when cultured in 2 % serum-containing medium. However, TSG treatment during NSC differentiation in vitro increased the number of Tuj-1-positive neurons, as well as the proportion of tyrosine hydroxylase(TH)-positive cells and dopamine- transporter- positive neurons, a late marker of mature DA neurons. We also found that TSG enhanced the expression of nuclear receptor related factor 1, a transcription factor specific for the development and maintenance of midbrain DA neurons in inducing NSC differentiation into TH -immunoreactive DA neurons. Moreover, TSG upregulated the expression of Wnt/ß-catenin signaling molecules (Wnt1, Wnt3a, Wnt5a, and ß-catenin). However, these promoting effects were significantly inhibited by the application of IWR1, a Wnt signaling-specific blocker in culture. Our findings suggested that TSG may have potential in inducing the DA neuronal differentiation of mouse NSCs mediated by triggering the Wnt/ß-catenin signaling pathway. These results indicated the possible role for TSG in the transplantation of NSCs for PD.


Subject(s)
Cell Differentiation/drug effects , Dopaminergic Neurons/drug effects , Glucosides/pharmacology , Mesencephalon/drug effects , Neural Stem Cells/drug effects , Stilbenes/pharmacology , Animals , Cell Differentiation/physiology , Cells, Cultured , Dopaminergic Neurons/physiology , Female , Glucosides/therapeutic use , Mesencephalon/cytology , Mesencephalon/physiology , Mice , Mice, Inbred BALB C , Neural Stem Cells/physiology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Pregnancy , Stilbenes/therapeutic use , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology
12.
J Cereb Blood Flow Metab ; 41(2): 197-205, 2021 02.
Article in English | MEDLINE | ID: mdl-33167744

ABSTRACT

At least two-thirds of spinal cord injury cases are anatomically incomplete, without complete spinal cord transection, although the initial injuries cause complete loss of sensory and motor functions. The malleability of neural circuits and networks allows varied extend of functional restoration in some individuals after successful rehabilitative training. However, in most cases, the efficiency and extent are both limited and uncertain, largely due to the many obstacles of repair. The restoration of function after anatomically incomplete injury is in part made possible by the growth of new axons or new axon branches through the spared spinal cord tissue and the new synaptic connections they make, either along the areas they grow through or in the areas they terminate. This review will discuss new progress on the understanding of the role of axon guidance molecules, particularly the Wnt family proteins, in spinal cord injury and how the knowledge and tools of axon guidance can be applied to increase the potential of recovery. These strategies, combined with others, such as neuroprotection and rehabilitation, may bring new promises. The recovery strategies for anatomically incomplete spinal cord injuries are relevant and may be applicable to traumatic brain injury and stroke.


Subject(s)
Axon Guidance/physiology , Nerve Regeneration/physiology , Spinal Cord Injuries/metabolism , Animals , Humans
13.
J Bacteriol ; 202(24)2020 11 19.
Article in English | MEDLINE | ID: mdl-32868406

ABSTRACT

Nucleotide-activated effector deployment, prototyped by interferon-dependent immunity, is a common mechanistic theme shared by immune systems of several animals and prokaryotes. Prokaryotic versions include CRISPR-Cas with the CRISPR polymerase domain, their minimal variants, and systems with second messenger oligonucleotide or dinucleotide synthetase (SMODS). Cyclic or linear oligonucleotide signals in these systems help set a threshold for the activation of potentially deleterious downstream effectors in response to invader detection. We establish such a regulatory mechanism to be a more general principle of immune systems, which can also operate independently of such messengers. Using sensitive sequence analysis and comparative genomics, we identify 12 new prokaryotic immune systems, which we unify by this principle of threshold-dependent effector activation. These display regulatory mechanisms paralleling physiological signaling based on 3'-5' cyclic mononucleotides, NAD+-derived messengers, two- and one-component signaling that includes histidine kinase-based signaling, and proteolytic activation. Furthermore, these systems allowed the identification of multiple new sensory signal sensory components, such as a tetratricopeptide repeat (TPR) scaffold predicted to recognize NAD+-derived signals, unreported versions of the STING domain, prokaryotic YEATS domains, and a predicted nucleotide sensor related to receiver domains. We also identify previously unrecognized invader detection components and effector components, such as prokaryotic versions of the Wnt domain. Finally, we show that there have been multiple acquisitions of unidentified STING domains in eukaryotes, while the TPR scaffold was incorporated into the animal immunity/apoptosis signal-regulating kinase (ASK) signalosome.IMPORTANCE Both prokaryotic and eukaryotic immune systems face the dangers of premature activation of effectors and degradation of self-molecules in the absence of an invader. To mitigate this, they have evolved threshold-setting regulatory mechanisms for the triggering of effectors only upon the detection of a sufficiently strong invader signal. This work defines general templates for such regulation in effector-based immune systems. Using this, we identify several previously uncharacterized prokaryotic immune mechanisms that accomplish the regulation of downstream effector deployment by using nucleotide, NAD+-derived, two-component, and one-component signals paralleling physiological homeostasis. This study has also helped identify several previously unknown sensor and effector modules in these systems. Our findings also augment the growing evidence for the emergence of key animal immunity and chromatin regulatory components from prokaryotic progenitors.


Subject(s)
Bacteria/genetics , Bacteria/immunology , Bacterial Proteins/immunology , Eukaryota/immunology , Amino Acid Sequence , Bacteria/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Eukaryota/genetics , Genomics , Immune System , Nucleotides/chemistry , Nucleotides/immunology , Sequence Alignment
14.
Front Oncol ; 10: 1341, 2020.
Article in English | MEDLINE | ID: mdl-32923386

ABSTRACT

Colorectal cancer (CRC) is the fourth leading cause of cancer death worldwide, and constitutive activation of the Wnt signaling pathway is universal in most CRC cases. Wnt ligands (Wnts) are secreted glycoproteins and fundamentally essential for the transduction of Wnt signaling pathway. However, the 19 members of Wnts in humans imply a daunting complexity of Wnt signaling and biological effects, and our understanding of their roles in CRC tumorigenesis is still quite rudimentary. This review will give an overview of the structural characteristics and maturation process of Wnts. The expression pattern of all human Wnts in CRC tissues, including Wnt1, Wnt2, Wnt2b, Wnt3, Wnt3a, Wnt4, Wnt5a, Wnt5b, Wnt6, Wnt7a, Wnt7b, Wnt8a, Wnt8b, Wnt9a, Wnt9b, Wnt10a, Wnt10b, Wnt11, and Wnt16, and their relationship with the tumorigenesis and the progression of CRC will be specifically summarized separately. Despite certain challenges, Wnt-based therapeutics for CRC emerge continuously and some are now in clinical trials. In conclusion, a deep understanding of Wnts is very helpful for a better management of this disease.

15.
Biochim Biophys Acta Mol Cell Res ; 1867(11): 118806, 2020 11.
Article in English | MEDLINE | ID: mdl-32739525

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a specific form of chronic, progressive and fibrosing interstitial pneumonia of unknown cause. The main feature of IPF is a heterogeneous appearance with areas of sub-pleural fibrosis. However, the mechanism of sub-pleural fibrosis was poorly understood. In this study, our in vivo study revealed that pleural mesothelial cells (PMCs) migrated into lung parenchyma and localized alongside lung fibroblasts in sub-pleural area in mouse pulmonary fibrosis. Our in vitro study displayed that cultured-PMCs-medium induced lung fibroblasts transforming into myofibroblast, cultured-fibroblasts-medium promoted mesothelial-mesenchymal transition of PMCs. Furthermore, these changes in lung fibroblasts and PMCs were prevented by blocking TGF-ß1/Smad2/3 signaling with SB431542. TGF-ß1 neutralized antibody attenuated bleomycin-induced pulmonary fibrosis. Similar to TGF-ß1/Smad2/3 signaling, wnt/ß-catenin signaling was also activated in the process of PMCs crosstalk with lung fibroblasts. Moreover, inhibition of CD147 attenuated cultured-PMCs-medium induced collagen-I synthesis in lung fibroblasts. Blocking CD147 signaling also prevented bleomycin-induced pulmonary fibrosis. Our data indicated that crosstalk between PMC and lung fibroblast contributed to sub-pleural pulmonary fibrosis. TGF-ß1, Wnt/ß-catenin and CD147 signaling was involved in the underling mechanism.


Subject(s)
Epithelium/drug effects , Lung/metabolism , Pleura/drug effects , Pulmonary Fibrosis/genetics , Animals , Benzamides/pharmacology , Cell Movement/genetics , Dioxoles/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelium/pathology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Lung/drug effects , Lung/pathology , Mice , Pleura/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Smad2 Protein/genetics , Transforming Growth Factor beta1/genetics
16.
Dev Biol ; 464(1): 88-102, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32473955

ABSTRACT

Cells in multicellular organisms rely on secreted ligands for development and morphogenesis. Several mechanisms modulate the availability and distribution of secreted ligands, determining their ability to signal locally and at long range from their source. One of these mechanisms is Dally-like protein (Dlp), a cell-surface glypican that exhibits biphasic functions in Drosophila wing discs, promoting Wg signaling at long-range from Wg source cells and inhibiting Wg signaling near source cells. In the germarium at the tip of the ovary, Dlp promotes long-range distribution of Wg from cap cells to follicle stem cells. However, the germarium also expresses other Wnts - Wnt2, Wnt4, and Wnt6 - that function locally in escort cells to promote oogenesis. Whether and how local functions of these Wnts are regulated remains unknown. Here we show that the dlp overexpression phenotype is multifaceted and phenocopies multiple Wnt loss-of-function phenotypes. Each aspect of dlp overexpression phenotype is suppressed by co-expression of individual Wnts, and the suppression pattern exhibited by each Wnt suggests that Wnts have functional specificity in the germarium. Further, dlp knockdown phenocopies Wnt gain-of-function phenotypes. Together these data show that Dlp inhibits the functions of each Wnt. All four Wnts co-immunoprecipitate with Dlp in S2R+ â€‹cells, suggesting that in the germarium, Dlp sequesters Wnts to inhibit local paracrine Wnt signaling. Our results indicate that Dlp modulates the availability of multiple extracellular Wnts for local paracrine Wnt signaling in the germarium.


Subject(s)
Drosophila Proteins/metabolism , Oogenesis/physiology , Ovary/metabolism , Paracrine Communication/physiology , Proteoglycans/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Ovary/cytology , Proteoglycans/genetics , Wnt Proteins/genetics
17.
Front Mol Neurosci ; 13: 86, 2020.
Article in English | MEDLINE | ID: mdl-32547365

ABSTRACT

Myasthenia gravis (MG) is a disease of the postsynaptic neuromuscular junction (NMJ) where nicotinic acetylcholine (ACh) receptors (AChRs) are targeted by autoantibodies. Search for other pathogenic antigens has detected the antibodies against muscle-specific tyrosine kinase (MuSK) and low-density lipoprotein-related protein 4 (Lrp4), both causing pre- and post-synaptic impairments. Agrin is also suspected as a fourth pathogen. In a complex NMJ organization centering on MuSK: (1) the Wnt non-canonical pathway through the Wnt-Lrp4-MuSK cysteine-rich domain (CRD)-Dishevelled (Dvl, scaffold protein) signaling acts to form AChR prepatterning with axonal guidance; (2) the neural agrin-Lrp4-MuSK (Ig1/2 domains) signaling acts to form rapsyn-anchored AChR clusters at the innervated stage of muscle; (3) adaptor protein Dok-7 acts on MuSK activation for AChR clustering from "inside" and also on cytoskeleton to stabilize AChR clusters by the downstream effector Sorbs1/2; (4) the trans-synaptic retrograde signaling contributes to the presynaptic organization via: (i) Wnt-MuSK CRD-Dvl-ß catenin-Slit 2 pathway; (ii) Lrp4; and (iii) laminins. The presynaptic Ca2+ homeostasis conditioning ACh release is modified by autoreceptors such as M1-type muscarinic AChR and A2A adenosine receptors. The post-synaptic structure is stabilized by: (i) laminin-network including the muscle-derived agrin; (ii) the extracellular matrix proteins (including collagen Q/perlecan and biglycan which link to MuSK Ig1 domain and CRD); and (iii) the dystrophin-associated glycoprotein complex. The study on MuSK ectodomains (Ig1/2 domains and CRD) recognized by antibodies suggested that the MuSK antibodies were pathologically heterogeneous due to their binding to multiple functional domains. Focussing one of the matrix proteins, biglycan which functions in the manner similar to collagen Q, our antibody assay showed the negative result in MG patients. However, the synaptic stability may be impaired by antibodies against MuSK ectodomains because of the linkage of biglycan with MuSK Ig1 domain and CRD. The pathogenic diversity of MG is discussed based on NMJ signaling molecules.

18.
Expert Opin Ther Targets ; 23(12): 1031-1039, 2019 12.
Article in English | MEDLINE | ID: mdl-31815563

ABSTRACT

Introduction: Osteoporotic fractures represent a growing burden of mortality, morbidity and socioeconomic cost to health-care systems worldwide. Osteoporosis is a disease uniquely associated with aging, therefore, an understanding of the physiological mechanisms underpinning its development as we age may open new avenues for therapeutic exploitation. Novel treatments, as well as refinement of the current approaches, are vital in the effort to sustain healthy, independent patients across the lifespan.Areas covered: This review covers the anabolic and catabolic pathways seen in bone maintenance, highlighting how they are changed with age, leading to osteoporosis. It will also discuss how these changes may be targeted therapeutically, in the development of new therapies, and the refinement of those already in use.Expert opinion: New effective and safe treatments for osteoporosis are still needed. Bone anabolics seem to be the most appropriate therapeutic approach to osteoporosis in older persons. Considering that bone and muscle mass synchronically decline with aging thus predisposing older persons to falls and fractures, combined therapeutic approaches to osteosarcopenia with a dual anabolic effect on muscle and bone will be a major advance in the treatment of these devastating conditions in the future.


Subject(s)
Bone Density Conservation Agents/pharmacology , Osteoporosis/drug therapy , Osteoporotic Fractures/prevention & control , Aged , Aging , Anabolic Agents/administration & dosage , Anabolic Agents/pharmacology , Animals , Bone Density Conservation Agents/administration & dosage , Bone and Bones/drug effects , Bone and Bones/metabolism , Drug Development , Humans , Osteoporosis/complications , Osteoporosis/physiopathology , Sarcopenia/drug therapy , Sarcopenia/physiopathology
19.
Int J Mol Sci ; 20(11)2019 Jun 08.
Article in English | MEDLINE | ID: mdl-31181747

ABSTRACT

Neuromuscular junction (NMJ) formation involves morphological changes both in motor terminals and muscle membrane. The molecular mechanisms leading to NMJ formation and maintenance have not yet been fully elucidated. During the last decade, it has become clear that virtually all cells release different types of extracellular vesicles (EVs), which can be taken up by nearby or distant cells modulating their activity. Initially, EVs were associated to a mechanism involved in the elimination of unwanted material; subsequent evidence demonstrated that exosomes, and more in general EVs, play a key role in intercellular communication by transferring proteins, lipids, DNA and RNA to target cells. Recently, EVs have emerged as potent carriers for Wnt, bone morphogenetic protein, miRNA secretion and extracellular traveling. Convincing evidence demonstrates that presynaptic terminals release exosomes that are taken up by muscle cells, and these exosomes can modulate synaptic plasticity in the recipient muscle cell in vivo. Furthermore, recent data highlighted that EVs could also be a potential cause of neurodegenerative disorders. Indeed, mutant SOD1, TDP-43 and FUS/TLS can be secreted by neural cells packaged into EVs and enter in neighboring neural cells, contributing to the onset and severity of the disease.


Subject(s)
Extracellular Vesicles/metabolism , Neuromuscular Junction/metabolism , Signal Transduction , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Motor Neuron Disease/etiology , Neurogenesis , Neuromuscular Junction/cytology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiology
20.
Cell ; 175(2): 372-386.e17, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30270042

ABSTRACT

Intestinal mesenchymal cells play essential roles in epithelial homeostasis, matrix remodeling, immunity, and inflammation. But the extent of heterogeneity within the colonic mesenchyme in these processes remains unknown. Using unbiased single-cell profiling of over 16,500 colonic mesenchymal cells, we reveal four subsets of fibroblasts expressing divergent transcriptional regulators and functional pathways, in addition to pericytes and myofibroblasts. We identified a niche population located in proximity to epithelial crypts expressing SOX6, F3 (CD142), and WNT genes essential for colonic epithelial stem cell function. In colitis, we observed dysregulation of this niche and emergence of an activated mesenchymal population. This subset expressed TNF superfamily member 14 (TNFSF14), fibroblastic reticular cell-associated genes, IL-33, and Lysyl oxidases. Further, it induced factors that impaired epithelial proliferation and maturation and contributed to oxidative stress and disease severity in vivo. Our work defines how the colonic mesenchyme remodels to fuel inflammation and barrier dysfunction in IBD.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Mesoderm/physiology , Animals , Cell Proliferation , Colitis/genetics , Colitis/physiopathology , Colon/physiology , Epithelial Cells/metabolism , Fibroblasts/physiology , Genetic Heterogeneity , Homeostasis , Humans , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/physiology , Intestines/immunology , Intestines/physiology , Mesenchymal Stem Cells/physiology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts , Pericytes , RAW 264.7 Cells , SOXD Transcription Factors/physiology , Single-Cell Analysis/methods , Thromboplastin/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Wnt Signaling Pathway/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...