Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 14.247
Filtrar
1.
J Clin Invest ; 134(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38949024

RESUMO

Mitochondria-related neurodegenerative diseases have been implicated in the disruption of primary cilia function. Mutation in an intrinsic mitochondrial complex I component NDUFAF2 has been identified in Leigh syndrome, a severe inherited mitochondriopathy. Mutations in ARMC9, which encodes a basal body protein, cause Joubert syndrome, a ciliopathy with defects in the brain, kidney, and eye. Here, we report a mechanistic link between mitochondria metabolism and primary cilia signaling. We discovered that loss of NDUFAF2 caused both mitochondrial and ciliary defects in vitro and in vivo and identified NDUFAF2 as a binding partner for ARMC9. We also found that NDUFAF2 was both necessary and sufficient for cilia formation and that exogenous expression of NDUFAF2 rescued the ciliary and mitochondrial defects observed in cells from patients with known ARMC9 deficiency. NAD+ supplementation restored mitochondrial and ciliary dysfunction in ARMC9-deficient cells and zebrafish and ameliorated the ocular motility and motor deficits of a patient with ARMC9 deficiency. The present results provide a compelling mechanistic link, supported by evidence from human studies, between primary cilia and mitochondrial signaling. Importantly, our findings have significant implications for the development of therapeutic approaches targeting ciliopathies.


Assuntos
Cílios , Doenças Renais Císticas , Doença de Leigh , Mitocôndrias , Peixe-Zebra , Humanos , Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Doença de Leigh/genética , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Cílios/metabolismo , Cílios/patologia , Cílios/genética , Animais , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitocôndrias/genética , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/genética , Proteínas do Domínio Armadillo/metabolismo , Proteínas do Domínio Armadillo/genética , Retina/metabolismo , Retina/patologia , Retina/anormalidades , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Anormalidades do Olho/metabolismo , Camundongos , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Cerebelo/metabolismo , Cerebelo/patologia , Cerebelo/anormalidades , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Masculino
2.
Cell Commun Signal ; 22(1): 348, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961488

RESUMO

BACKGROUND: Primary cilia on the surface of eukaryotic cells serve as sensory antennas for the reception and transmission in various cell signaling pathways. They are dynamic organelles that rapidly form during differentiation and cell cycle exit. Defects in these organelles cause a group of wide-ranging disorders called ciliopathies. Tonicity-responsive enhancer-binding protein (TonEBP) is a pleiotropic stress protein that mediates various physiological and pathological cellular responses. TonEBP is well-known for its role in adaptation to a hypertonic environment, to which primary cilia have been reported to contribute. Furthermore, TonEBP is involved in a wide variety of other signaling pathways, such as Sonic Hedgehog and WNT signaling, that promote primary ciliogenesis, suggesting a possible regulatory role. However, the functional relationship between TonEBP and primary ciliary formation remains unclear. METHODS: TonEBP siRNAs and TonEBP-mCherry plasmids were used to examine their effects on cell ciliation rates, assembly and disassembly processes, and regulators. Serum starvation was used as a condition to induce ciliogenesis. RESULTS: We identified a novel pericentriolar localization for TonEBP. The results showed that TonEBP depletion facilitates the formation of primary cilia, whereas its overexpression results in fewer ciliated cells. Moreover, TonEBP controlled the expression and activity of aurora kinase A, a major negative regulator of ciliogenesis. Additionally, TonEBP overexpression inhibited the loss of CP110 from the mother centrioles during the early stages of primary cilia assembly. Finally, TonEBP regulated the localization of PCM1 and AZI1, which are necessary for primary cilia formation. CONCLUSIONS: This study proposes a novel role for TonEBP as a pericentriolar protein that regulates the integrity of centriolar satellite components. This regulation has shown to have a negative effect on ciliogenesis. Investigations into cilium assembly and disassembly processes suggest that TonEBP acts upstream of the aurora kinase A - histone deacetylase 6 signaling pathway and affects basal body formation to control ciliogenesis. Taken together, our data proposes previously uncharacterized regulation of primary cilia assembly by TonEBP.


Assuntos
Aurora Quinase A , Centríolos , Cílios , Cílios/metabolismo , Humanos , Aurora Quinase A/metabolismo , Aurora Quinase A/genética , Centríolos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Desacetilase 6 de Histona/metabolismo , Desacetilase 6 de Histona/genética , Animais , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/genética
3.
Cells ; 13(13)2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38995007

RESUMO

Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.


Assuntos
Cílios , Células-Tronco Pluripotentes Induzidas , Proteínas de Membrana , Cílios/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Neurônios/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Transporte Proteico
4.
Proc Natl Acad Sci U S A ; 121(28): e2320070121, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38968120

RESUMO

Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.


Assuntos
Quinases Dyrk , Proteínas Hedgehog , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Transdução de Sinais , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco , Animais , Proteína Gli3 com Dedos de Zinco/metabolismo , Proteína Gli3 com Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Camundongos , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Humanos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Proliferação de Células , Cílios/metabolismo , Receptor Smoothened/metabolismo , Receptor Smoothened/genética , Proteínas Nucleares , Proteínas Repressoras
5.
Stem Cell Res Ther ; 15(1): 198, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38971766

RESUMO

BACKGROUND: Trans-sutural distraction osteogenesis (TSDO) involves the application of distraction force to facial sutures to stimulate osteogenesis. Gli1+ cells in the cranial sutures play an important role in bone growth. However, whether Gli1+ cells in facial sutures differentiate into bone under distraction force is unknown. METHODS: 4-week-old Gli1ER/Td and C57BL/6 mice were used to establish a TSDO model to explore osteogenesis of zygomaticomaxillary sutures. A Gli1+ cell lineage tracing model was used to observe the distribution of Gli1+ cells and explore the role of Gli1+ cells in facial bone remodeling. RESULTS: Distraction force promoted bone remodeling during TSDO. Fluorescence and two-photon scanning images revealed the distribution of Gli1+ cells. Under distraction force, Gli1-lineage cells proliferated significantly and co-localized with Runx2+ cells. Hedgehog signaling was upregulated in Gli1+ cells. Inhibition of Hedgehog signaling suppresses the proliferation and osteogenesis of Gli1+ cells induced by distraction force. Subsequently, the stem cell characteristics of Gli1+ cells were identified. Cell-stretching experiments verified that mechanical force promoted the osteogenic differentiation of Gli1+ cells through Hh signaling. Furthermore, immunofluorescence staining and RT-qPCR experiments demonstrated that the primary cilia in Gli1+ cells exhibit Hedgehog-independent mechanosensitivity, which was required for the osteogenic differentiation induced by mechanical force. CONCLUSIONS: Our study indicates that the primary cilia of Gli1+ cells sense mechanical stimuli, mediate Hedgehog signaling activation, and promote the osteogenic differentiation of Gli1+ cells in zygomaticomaxillary sutures.


Assuntos
Diferenciação Celular , Cílios , Suturas Cranianas , Proteínas Hedgehog , Osteogênese , Transdução de Sinais , Proteína GLI1 em Dedos de Zinco , Animais , Camundongos , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Osteogênese/fisiologia , Cílios/metabolismo , Suturas Cranianas/metabolismo , Camundongos Endogâmicos C57BL , Osteogênese por Distração/métodos , Proliferação de Células
6.
Biol Pharm Bull ; 47(7): 1307-1313, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39019610

RESUMO

Airway ciliary cells are components of the mucociliary transport system and play an important role in sweeping out small particles, such as bacteria and viruses, towards the oropharynx by the action of beating cilia. Several lines of evidence have shown that the ciliary beat is under the regulation of the purinergic system; however, the subtype of receptor and the intracellular signaling pathways involved in the activation of ciliary movement remain to be elucidated. In addition, although the activity of ciliary movement comprises two parameters, the ciliary beat frequency (CBF) and ciliary bend angle (CBA), few reports have analyzed CBA. In this study, we examined the effects of ATP and other purinergic ligands on both CBF and CBA and demonstrated that the purinergic signaling requirements for CBF and CBA are different, with CBF mediated by P2Y1 receptor activation and CBA mediated by the P2X4 receptor.


Assuntos
Trifosfato de Adenosina , Brônquios , Cílios , Animais , Cílios/metabolismo , Cílios/fisiologia , Trifosfato de Adenosina/metabolismo , Camundongos , Brônquios/citologia , Depuração Mucociliar/fisiologia , Masculino , Receptores Purinérgicos P2X4/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos/metabolismo , Transdução de Sinais
7.
Proc Natl Acad Sci U S A ; 121(31): e2320303121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39008691

RESUMO

Influenza viruses pose a significant burden on global human health. Influenza has a broad cellular tropism in the airway, but how infection of different epithelial cell types impacts replication kinetics and burden in the airways is not fully understood. Using primary human airway cultures, which recapitulate the diverse epithelial cell landscape of the human airways, we investigated the impact of cell type composition on virus tropism and replication kinetics. Cultures were highly diverse across multiple donors and 30 independent differentiation conditions and supported a range of influenza replication. Although many cell types were susceptible to influenza, ciliated and secretory cells were predominantly infected. Despite the strong tropism preference for secretory and ciliated cells, which consistently make up 75% or more of infected cells, only ciliated cells were associated with increased virus production. Surprisingly, infected secretory cells were associated with overall reduced virus output. The disparate response and contribution to influenza virus production could be due to different pro- and antiviral interferon-stimulated gene signatures between ciliated and secretory populations, which were interrogated with single-cell RNA sequencing. These data highlight the heterogeneous outcomes of influenza virus infections in the complex cellular environment of the human airway and the disparate impacts of infected cell identity on multiround burst size, even among preferentially infected cell types.


Assuntos
Células Epiteliais , Influenza Humana , Tropismo Viral , Replicação Viral , Humanos , Influenza Humana/virologia , Replicação Viral/fisiologia , Células Epiteliais/virologia , Células Epiteliais/metabolismo , Cílios/virologia , Cílios/metabolismo , Células Cultivadas , Mucosa Respiratória/virologia , Mucosa Respiratória/citologia
10.
Development ; 151(14)2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39007638

RESUMO

Vertebrate motile cilia are classified as (9+2) or (9+0), based on the presence or absence of the central pair apparatus, respectively. Cryogenic electron microscopy analyses of (9+2) cilia have uncovered an elaborate axonemal protein composition. The extent to which these features are conserved in (9+0) cilia remains unclear. CFAP53, a key axonemal filamentous microtubule inner protein (fMIP) and a centriolar satellites component, is essential for motility of (9+0), but not (9+2) cilia. Here, we show that in (9+2) cilia, CFAP53 functions redundantly with a paralogous fMIP, MNS1. MNS1 localises to ciliary axonemes, and combined loss of both proteins in zebrafish and mice caused severe outer dynein arm loss from (9+2) cilia, significantly affecting their motility. Using immunoprecipitation, we demonstrate that, whereas MNS1 can associate with itself and CFAP53, CFAP53 is unable to self-associate. We also show that additional axonemal dynein-interacting proteins, two outer dynein arm docking (ODAD) complex members, show differential localisation between types of motile cilia. Together, our findings clarify how paralogous fMIPs, CFAP53 and MNS1, function in regulating (9+2) versus (9+0) cilia motility, and further emphasise extensive structural diversity among these organelles.


Assuntos
Axonema , Cílios , Peixe-Zebra , Animais , Cílios/metabolismo , Cílios/ultraestrutura , Peixe-Zebra/metabolismo , Camundongos , Axonema/metabolismo , Axonema/ultraestrutura , Dineínas do Axonema/metabolismo , Dineínas do Axonema/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Dineínas/metabolismo
11.
Int J Mol Sci ; 25(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38892227

RESUMO

The primary cilium is a microtubule-based sensory organelle that plays a critical role in signaling pathways and cell cycle progression. Defects in the structure and/or function of the primary cilium result in developmental diseases collectively known as ciliopathies. However, the constituents and regulatory mechanisms of the primary cilium are not fully understood. In recent years, the activity of the epigenetic modifier SMYD3 has been shown to play a key role in the regulation of cell cycle progression. However, whether SMYD3, a histone/lysine methyltransferase, contributes to the regulation of ciliogenesis remains unknown. Here, we report that SMYD3 drives ciliogenesis via the direct and indirect regulation of cilia-associated components. We show that SMYD3 is a novel component of the distal appendage and is required for centriolar appendage assembly. The loss of SMYD3 decreased the percentage of ciliated cells and resulted in the formation of stumpy cilia. We demonstrated that SMYD3 modulated the recruitment of centrosome proteins (Cep164, Fbf1, Ninein, Ttbk2 and Cp110) and the trafficking of intraflagellar transport proteins (Ift54 and Ift140) important for cilia formation and maintenance, respectively. In addition, we showed that SMYD3 regulated the transcription of cilia genes and bound to the promoter regions of C2cd3, Cep164, Ttbk2, Dync2h1 and Cp110. This study provides insights into the role of SMYD3 in cilia biology and suggests that SMYD3-mediated cilia formation/function may be relevant for cilia-dependent signaling in ciliopathies.


Assuntos
Centrossomo , Cílios , Histona-Lisina N-Metiltransferase , Transporte Proteico , Cílios/metabolismo , Humanos , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Centrossomo/metabolismo , Animais , Flagelos/metabolismo , Camundongos , Proteínas Associadas a Centrossomos
12.
J Cell Sci ; 137(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38853670

RESUMO

Intraflagellar transport (IFT) is required for ciliary assembly. The IFT machinery comprises the IFT motors kinesin-2 and IFT dynein plus IFT-A and IFT-B complexes, which assemble into IFT trains in cilia. To gain mechanistic understanding of IFT and ciliary assembly, here, we performed an absolute quantification of IFT machinery in Chlamydomonas reinhardtii cilium. There are ∼756, ∼532, ∼276 and ∼350 molecules of IFT-B, IFT-A, IFT dynein and kinesin-2, respectively, per cilium. The amount of IFT-B is sufficient to sustain rapid ciliary growth in terms of tubulin delivery. The stoichiometric ratio of IFT-B:IFT-A:dynein is ∼3:2:1 whereas the IFT-B:IFT-A ratio in an IFT dynein mutant is 2:1, suggesting that there is a plastic interaction between IFT-A and IFT-B that can be influenced by IFT dynein. Considering diffusion of kinesin-2 during retrograde IFT, it is estimated that one kinesin-2 molecule drives eight molecules of IFT-B during anterograde IFT. These data provide new insights into the assembly of IFT trains and ciliary assembly.


Assuntos
Chlamydomonas reinhardtii , Cílios , Dineínas , Flagelos , Cinesinas , Proteômica , Cílios/metabolismo , Chlamydomonas reinhardtii/metabolismo , Chlamydomonas reinhardtii/genética , Proteômica/métodos , Cinesinas/metabolismo , Cinesinas/genética , Dineínas/metabolismo , Flagelos/metabolismo , Transporte Biológico
13.
Cells ; 13(12)2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38920647

RESUMO

Defects in motile cilia, termed motile ciliopathies, result in clinical manifestations affecting the respiratory and reproductive system, as well as laterality defects and hydrocephalus. We previously defined biallelic MNS1 variants causing situs inversus and male infertility, mirroring the findings in Mns1-/- mice. Here, we present clinical and genomic findings in five newly identified individuals from four unrelated families affected by MNS1-related disorder. Ciliopathy panel testing and whole exome sequencing identified one previously reported and two novel MNS1 variants extending the genotypic spectrum of disease. A broad spectrum of laterality defects including situs inversus totalis and heterotaxia was confirmed. Interestingly, a single affected six-year-old girl homozygous for an MNS1 nonsense variant presented with a history of neonatal respiratory distress syndrome, recurrent respiratory tract infections, chronic rhinitis, and wet cough. Accordingly, immunofluorescence analysis showed the absence of MNS1 from the respiratory epithelial cells of this individual. Two other individuals with hypomorphic variants showed laterality defects and mild respiratory phenotype. This study represents the first observation of heterotaxia and respiratory disease in individuals with biallelic MNS1 variants, an important extension of the phenotype associated with MNS1-related motile ciliopathy disorder.


Assuntos
Alelos , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Cílios/patologia , Cílios/genética , Ciliopatias/genética , Ciliopatias/patologia , Linhagem , Fenótipo , Lactente , Adolescente
14.
J Cell Sci ; 137(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38841887

RESUMO

Centrosomal proteins play pivotal roles in orchestrating microtubule dynamics, and their dysregulation leads to disorders, including cancer and ciliopathies. Understanding the multifaceted roles of centrosomal proteins is vital to comprehend their involvement in disease development. Here, we report novel cellular functions of CEP41, a centrosomal and ciliary protein implicated in Joubert syndrome. We show that CEP41 is an essential microtubule-associated protein with microtubule-stabilizing activity. Purified CEP41 binds to preformed microtubules, promotes microtubule nucleation and suppresses microtubule disassembly. When overexpressed in cultured cells, CEP41 localizes to microtubules and promotes microtubule bundling. Conversely, shRNA-mediated knockdown of CEP41 disrupts the interphase microtubule network and delays microtubule reassembly, emphasizing its role in microtubule organization. Further, we demonstrate that the association of CEP41 with microtubules relies on its conserved rhodanese homology domain (RHOD) and the N-terminal region. Interestingly, a disease-causing mutation in the RHOD domain impairs CEP41-microtubule interaction. Moreover, depletion of CEP41 inhibits cell proliferation and disrupts cell cycle progression, suggesting its potential involvement in cell cycle regulation. These insights into the cellular functions of CEP41 hold promise for unraveling the impact of its mutations in ciliopathies.


Assuntos
Proliferação de Células , Microtúbulos , Humanos , Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Centrossomo/metabolismo , Retina/metabolismo , Retina/patologia , Retina/anormalidades , Ciliopatias/metabolismo , Ciliopatias/genética , Ciliopatias/patologia , Cerebelo/metabolismo , Cerebelo/anormalidades , Cerebelo/patologia , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Cílios/metabolismo , Cílios/patologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Animais , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Anormalidades do Olho/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Ligação Proteica , Ciclo Celular/genética , Células HEK293
15.
Cells ; 13(12)2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38920633

RESUMO

The primary cilium, an antenna-like sensory organelle that protrudes from the surface of most eukaryotic cell types, has become a signaling hub of growing interest given that defects in its structure and/or function are associated with human diseases and syndromes, known as ciliopathies. With the continuously expanding role of primary cilia in health and diseases, identifying new players in ciliogenesis will lead to a better understanding of the function of this organelle. It has been shown that the primary cilium shares similarities with the immune synapse, a highly organized structure at the interface between an antigen-presenting or target cell and a lymphocyte. Studies have demonstrated a role for known cilia regulators in immune synapse formation. However, whether immune synapse regulators modulate ciliogenesis remains elusive. Here, we find that programmed death ligand 1 (PD-L1), an immune checkpoint protein and regulator of immune synapse formation, plays a role in the regulation of ciliogenesis. We found that PD-L1 is enriched at the centrosome/basal body and Golgi apparatus of ciliated cells and depleting PD-L1 enhanced ciliogenesis and increased the accumulation of ciliary membrane trafficking proteins Rab8a, BBS5, and sensory receptor protein PC-2. Moreover, PD-L1 formed a complex with BBS5 and PC-2. In addition, we found that depletion of PD-L1 resulted in the ciliary accumulation of Gli3 and the downregulation of Gli1. Our results suggest that PD-L1 is a new player in ciliogenesis, contributing to PC-2-mediated sensory signaling and the Hh signaling cascade.


Assuntos
Antígeno B7-H1 , Cílios , Proteínas Hedgehog , Transdução de Sinais , Cílios/metabolismo , Antígeno B7-H1/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Animais , Camundongos , Centrossomo/metabolismo , Complexo de Golgi/metabolismo
16.
Development ; 151(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38940293

RESUMO

Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.


Assuntos
Diferenciação Celular , Hematopoese , Células-Tronco Hematopoéticas , Peixe-Zebra , Animais , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Diferenciação Celular/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Corpos Embrioides/citologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Cílios/metabolismo , Cílios/efeitos dos fármacos , Blastômeros/citologia , Blastômeros/metabolismo , Blastômeros/efeitos dos fármacos , Células Cultivadas
17.
PLoS Pathog ; 20(6): e1012303, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38885287

RESUMO

Chlamydia trachomatis is a clinically important bacterium that infects epithelial cells of the genitourinary and respiratory tracts and the eye. These differentiated cells are in a quiescent growth state and have a surface organelle called a primary cilium, but the standard Chlamydia cell culture infection model uses cycling cells that lack primary cilia. To investigate if these differences are relevant, we performed infections with host cells that have a primary cilium. We found that C. trachomatis caused progressive loss of the primary cilium that was prevented by disrupting Aurora A (AurA), HDAC6 or calmodulin, which are components of the cellular cilia disassembly pathway. Stabilization of the primary cilium by targeting this pathway caused a large reduction in infectious progeny although there were no changes in chlamydial inclusion growth, chlamydial replication or the ultrastructural appearance of dividing and infectious forms (RBs and EBs, respectively). Thus, the presence of a primary cilium interfered with the production of infectious EBs at a late step in the developmental cycle. C. trachomatis infection also induced quiescent cells to re-enter the cell cycle, as detected by EdU incorporation in S-phase, and Chlamydia-induced cilia disassembly was necessary for cell cycle re-entry. This study therefore describes a novel host-pathogen interaction in which the primary cilium limits a productive Chlamydia infection, and the bacterium counteracts this host cell defense by activating the cellular cilia disassembly pathway.


Assuntos
Infecções por Chlamydia , Chlamydia trachomatis , Cílios , Chlamydia trachomatis/fisiologia , Cílios/microbiologia , Cílios/metabolismo , Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/patologia , Humanos , Células Epiteliais/microbiologia , Células Epiteliais/metabolismo
18.
Phys Rev E ; 109(5-1): 054407, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38907471

RESUMO

Cilia are hairlike microactuators whose cyclic motion is specialized to propel extracellular fluids at low Reynolds numbers. Clusters of these organelles can form synchronized beating patterns, called metachronal waves, which presumably arise from hydrodynamic interactions. We model hydrodynamically interacting cilia by microspheres elastically bound to circular orbits, whose inclinations with respect to a no-slip wall model the ciliary power and recovery stroke, resulting in an anisotropy of the viscous flow. We derive a coupled phase-oscillator description by reducing the microsphere dynamics to the slow timescale of synchronization and determine analytical metachronal wave solutions and their stability in a periodic chain setting. In this framework, a simple intuition for the hydrodynamic coupling between phase oscillators is established by relating the geometry of flow near the surface of a cell or tissue to the directionality of the hydrodynamic coupling functions. This intuition naturally explains the properties of the linear stability of metachronal waves. The flow near the surface stabilizes metachronal waves with long wavelengths propagating in the direction of the power stroke and, moreover, metachronal waves with short wavelengths propagating perpendicularly to the power stroke. Performing simulations of phase-oscillator chains with periodic boundary conditions, we indeed find that both wave types emerge with a variety of linearly stable wave numbers. In open chains of phase oscillators, the dynamics of metachronal waves is fundamentally different. Here the elasticity of the model cilia controls the wave direction and selects a particular wave number: At large elasticity, waves traveling in the direction of the power stroke are stable, whereas at smaller elasticity waves in the opposite direction are stable. For intermediate elasticity both wave directions coexist. In this regime, waves propagating towards both ends of the chain form, but only one wave direction prevails, depending on the elasticity and initial conditions.


Assuntos
Cílios , Elasticidade , Hidrodinâmica , Modelos Biológicos , Cílios/fisiologia , Propriedades de Superfície , Microesferas
19.
Biol Pharm Bull ; 47(6): 1113-1118, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38839362

RESUMO

Motile cilia in the ependymal cells that line the brain ventricles play pivotal roles in cerebrospinal fluid (CSF) flow in well-defined directions. However, the substances and pathways which regulate their beating have not been well studied. Here, we used primary cultured cells derived from neonatal mouse brain that possess motile cilia and found that adenosine (ADO) stimulates ciliary beating by increasing the ciliary beat frequency (CBF) in a concentration-dependent manner, with the ED50 value being 5 µM. Ciliary beating stimulated by ADO was inhibited by A2B receptor (A2BR) antagonist MRS1754 without any inhibition by antagonists of other ADO receptor subtypes. The expression of A2BR on the cilia was also confirmed by immunofluorescence. The values of CBF were also increased by forskolin, which is an activator of adenylate cyclase, whereas they were not further increased by the addition of ADO. Furthermore, ciliary beating was not stimulated by ADO in the presence of a protein kinase A (PKA) inhibitors. These results altogether suggest that ADO stimulates ciliary beating through A2BR on the cilia, and activation of PKA.


Assuntos
Adenosina , Animais Recém-Nascidos , Encéfalo , Cílios , Proteínas Quinases Dependentes de AMP Cíclico , Receptor A2B de Adenosina , Animais , Cílios/efeitos dos fármacos , Cílios/metabolismo , Cílios/fisiologia , Receptor A2B de Adenosina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Adenosina/farmacologia , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Camundongos , Células Cultivadas , Transdução de Sinais/efeitos dos fármacos , Antagonistas do Receptor A2 de Adenosina/farmacologia , Colforsina/farmacologia , Epêndima/metabolismo , Epêndima/citologia
20.
J Cell Biol ; 223(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-38856684

RESUMO

Sonic Hedgehog (SHH) is a driver of embryonic patterning that, when corrupted, triggers developmental disorders and cancers. SHH effector responses are organized through primary cilia (PC) that grow and retract with the cell cycle and in response to extracellular cues. Disruption of PC homeostasis corrupts SHH regulation, placing significant pressure on the pathway to maintain ciliary fitness. Mechanisms by which ciliary robustness is ensured in SHH-stimulated cells are not yet known. Herein, we reveal a crosstalk circuit induced by SHH activation of Phospholipase A2α that drives ciliary E-type prostanoid receptor 4 (EP4) signaling to ensure PC function and stabilize ciliary length. We demonstrate that blockade of SHH-EP4 crosstalk destabilizes PC cyclic AMP (cAMP) equilibrium, slows ciliary transport, reduces ciliary length, and attenuates SHH pathway induction. Accordingly, Ep4-/- mice display shortened neuroepithelial PC and altered SHH-dependent neuronal cell fate specification. Thus, SHH initiates coordination between distinct ciliary receptors to maintain PC function and length homeostasis for robust downstream signaling.


Assuntos
Cílios , Proteínas Hedgehog , Prostaglandinas , Transdução de Sinais , Animais , Camundongos , Cílios/metabolismo , AMP Cíclico/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Camundongos Knockout , Prostaglandinas/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA