Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(15)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39120335

RESUMEN

The endosomal sorting complex required for transport (ESCRT) machinery is composed of an articulated architecture of proteins that assemble at multiple cellular sites. The ESCRT machinery is involved in pathways that are pivotal for the physiology of the cell, including vesicle transport, cell division, and membrane repair. The subunits of the ESCRT I complex are mainly responsible for anchoring the machinery to the action site. The ESCRT II subunits function to bridge and recruit the ESCRT III subunits. The latter are responsible for finalizing operations that, independently of the action site, involve the repair and fusion of membrane edges. In this review, we report on the data related to the activity of the ESCRT machinery at two sites: the nuclear membrane and the midbody and the bridge linking cells in the final stages of cytokinesis. In these contexts, the machinery plays a significant role for the protection of genome integrity by contributing to the control of the abscission checkpoint and to nuclear envelope reorganization and correlated resilience. Consistently, several studies show how the dysfunction of the ESCRT machinery causes genome damage and is a codriver of pathologies, such as laminopathies and cancer.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Humanos , Citocinesis , Animales , Membrana Nuclear/metabolismo , Inestabilidad Genómica
2.
Genetics ; 227(4)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38884207

RESUMEN

Extracellular vesicles (EVs) encompass a diverse array of membrane-bound organelles released outside cells in response to developmental and physiological cell needs. EVs play important roles in remodeling the shape and content of differentiating cells and can rescue damaged cells from toxic or dysfunctional content. EVs can send signals and transfer metabolites between tissues and organisms to regulate development, respond to stress or tissue damage, or alter mating behaviors. While many EV functions have been uncovered by characterizing ex vivo EVs isolated from body fluids and cultured cells, research using the nematode Caenorhabditis elegans has provided insights into the in vivo functions, biogenesis, and uptake pathways. The C. elegans EV field has also developed methods to analyze endogenous EVs within the organismal context of development and adult physiology in free-living, behaving animals. In this review, we summarize major themes that have emerged for C. elegans EVs and their relevance to human health and disease. We also highlight the diversity of biogenesis mechanisms, locations, and functions of worm EVs and discuss open questions and unexplored topics tenable in C. elegans, given the nematode model is ideal for light and electron microscopy, genetic screens, genome engineering, and high-throughput omics.


Asunto(s)
Caenorhabditis elegans , Vesículas Extracelulares , Animales , Vesículas Extracelulares/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Humanos
3.
Protein Cell ; 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801297

RESUMEN

Cytokinesis is required for faithful division of cytoplasmic components and duplicated nuclei into two daughter cells. Midbody, a protein-dense organelle that forms at the intercellular bridge, is indispensable for successful cytokinesis. However, the regulatory mechanism of cytokinesis at the midbody still remains elusive. Here, we unveil a critical role for NudC-like protein 2 (NudCL2), a co-chaperone of heat shock protein 90 (Hsp90), in cytokinesis regulation by stabilizing regulator of chromosome condensation 2 (RCC2) at the midbody in mammalian cells. NudCL2 localizes at the midbody, and its downregulation results in cytokinesis failure, multinucleation and midbody disorganization. Using iTRAQ-based quantitative proteomic analysis, we find that RCC2 levels are decreased in NudCL2 knockout (KO) cells. Moreover, Hsp90 forms a complex with NudCL2 to stabilize RCC2, which is essential for cytokinesis. RCC2 depletion mirrors phenotypes observed in NudCL2-downregulated cells. Importantly, ectopic expression of RCC2 rescues the cytokinesis defects induced by NudCL2 deletion, but not vice versa. Together, our data reveal the significance of the NudCL2/Hsp90/RCC2 pathway in cytokinesis at the midbody.

4.
Proteomics ; 24(11): e2300058, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38470197

RESUMEN

Previously, we reported that human primary (SW480) and metastatic (SW620) colorectal (CRC) cells release three classes of membrane-encapsulated extracellular vesicles (EVs); midbody remnants (MBRs), exosomes (Exos), and microparticles (MPs). We reported that MBRs were molecularly distinct at the protein level. To gain further biochemical insights into MBRs, Exos, and MPs and their emerging role in CRC, we performed, and report here, for the first time, a comprehensive transcriptome and long noncoding RNA sequencing analysis and fusion gene identification of these three EV classes using the next-generation RNA sequencing technique. Differential transcript expression analysis revealed that MBRs have a distinct transcriptomic profile compared to Exos and MPs with a high enrichment of mitochondrial transcripts lncRNA/pseudogene transcripts that are predicted to bind to ribonucleoprotein complexes, spliceosome, and RNA/stress granule proteins. A salient finding from this study is a high enrichment of several fusion genes in MBRs compared to Exos, MPs, and cell lysates from their parental cells such as MSH2 (gene encoded DNA mismatch repair protein MSH2). This suggests potential EV-liquid biopsy targets for cancer detection. Importantly, the expression of cancer progression-related transcripts found in EV classes derived from SW480 (EGFR) and SW620 (MET and MACCA1) cell lines reflects their parental cell types. Our study is the report of RNA and fusion gene compositions within MBRs (including Exos and MPs) that could have an impact on EV functionality in cancer progression and detection using EV-based RNA/ fusion gene candidates for cancer biomarkers.


Asunto(s)
Neoplasias Colorrectales , Exosomas , Perfilación de la Expresión Génica , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Exosomas/genética , Exosomas/metabolismo , Perfilación de la Expresión Génica/métodos , Línea Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Transcriptoma/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(2): 261-268, 2024 Apr 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38413234

RESUMEN

Recent studies have shown that the formation of the primary cilium is associated with a specific cellular organelle known as the midbody remnant (MBR), which is a point-like organelle formed by shedding of the midbody at the end of mitosis. MBRs move along the cell surface close to the center body and regulate it to form primary cilia at the top of the centriole. Primary cilia can act as an organelle to inhibit tumorigenesis, and it is lost in a variety of tumors. Studies have shown that the accumulation of MBRs in tumor cells affects ciliogenesis; in addition, both MBRs and primary cilia are degraded in tumor cells through the autophagy pathway, and MBRs can also transfer tumor signaling pathway factors to primary cilia affecting tumorigenesis. In this article, the basic structure and the formation process of MBR and primary cilia are reviewed and the mechanism of MBRs regulating ciliogenesis is elaborated. The significance of MBR-mediated ciliogenesis in tumorigenesis and its potential as a target for cancer treatment are discussed.


Asunto(s)
Cilios , Neoplasias , Cilios/fisiología , Cilios/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Autofagia/fisiología , Carcinogénesis , Centriolos/metabolismo , Centriolos/fisiología , Transducción de Señal , Orgánulos/metabolismo , Mitosis , Animales
6.
Cell Mol Life Sci ; 80(12): 365, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37982870

RESUMEN

The aim of this review article is to focus on the unconventional roles of epigenetic players (chromatin remodelers and long non-coding RNAs) in cell division, beyond their well-characterized functions in chromatin regulation during cell differentiation and development. In the last two  decades, diverse experimental evidence has shown that subunits of SRCAP and p400/TIP60 chromatin remodeling complexes in humans relocate from interphase nuclei to centrosomes, spindle or midbody, with their depletion yielding an array of aberrant outcomes of mitosis and cytokinesis. Remarkably, this behavior is shared by orthologous subunits of the Drosophila melanogaster DOM/TIP60 complex, despite fruit flies and humans diverged over 700 million years ago. In short, the available data support the view that subunits of these complexes are a new class of moonlighting proteins, in that they lead a "double life": during the interphase, they function in chromatin regulation within the nucleus, but as the cell progresses through mitosis, they interact with established mitotic factors, thus becoming integral components of the cell division apparatus.  By doing so, they contribute to ensuring the correct distribution of chromosomes in the two daughter cells and, when dysfunctional, can cause genomic instability, a condition that can trigger tumorigenesis and developmental diseases. Research over the past few years has unveiled a major contribution of long non-coding RNAs (lncRNAs) in the epigenetics regulation of gene expression which also impacts on cell division control. Here, we focus on possible structural roles of lncRNAs in the execution of cytokinesis: in particular, we suggest that specific classes of lncRNAs relocate to the midbody to form an architectural scaffold ensuring its proper assembly and function during abscission. Drawing attention to experimental evidence for non-canonical extranuclear roles of chromatin factors and lncRNAs has direct implications on important and novel aspects concerning both the epigenetic regulation and the evolutionary dynamics of cell division with a significant impact on differentiation, development, and diseases.


Asunto(s)
Cromatina , ARN Largo no Codificante , Humanos , Animales , Cromatina/genética , ARN Largo no Codificante/genética , Drosophila melanogaster/genética , Epigénesis Genética , Mitosis/genética , Drosophila
7.
Dev Cell ; 58(19): 1917-1932.e6, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37552987

RESUMEN

Long ignored as a vestigial remnant of cytokinesis, the mammalian midbody (MB) is released post-abscission inside large extracellular vesicles called MB remnants (MBRs). Recent evidence suggests that MBRs can modulate cell proliferation and cell fate decisions. Here, we demonstrate that the MB matrix is the site of ribonucleoprotein assembly and is enriched in mRNAs that encode proteins involved in cell fate, oncogenesis, and pluripotency, which we are calling the MB granule. Both MBs and post-abscission MBRs are sites of spatiotemporally regulated translation, which is initiated when nascent daughter cells re-enter G1 and continues after extracellular release. MKLP1 and ARC are necessary for the localization and translation of RNA in the MB dark zone, whereas ESCRT-III is necessary to maintain translation levels in the MB. Our work reveals a unique translation event that occurs during abscission and within a large extracellular vesicle.


Asunto(s)
Citocinesis , ARN , Animales , Humanos , Diferenciación Celular , Células HeLa , Mamíferos
8.
FEBS J ; 290(24): 5704-5719, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37549045

RESUMEN

YES-associated protein (YAP) is a part of the Hippo pathway, with pivotal roles in several developmental processes and dual functionality as both a tumor suppressor and an oncogene. In the present study, we identified YAP activity as a microtubular scaffold protein that maintains the stability of the mitotic spindle and midbody by physically interacting with α-tubulin during mitotic progression. The interaction of YAP and α-tubulin was evident in co-immunoprecipitation assays, as well as observing their co-localization in the microtubular structure of the mitotic spindle and midbody in immunostainings. With YAP depletion, levels of ECT2, MKLP-1, and Aurora B are reduced, which is consistent with YAP functioning in midbody formation during cytokinesis. The concomitant decrease in α-tubulin and increase in acetyl-α-tubulin during YAP depletion occurred at the post-transcriptional level. This suggests that YAP maintains the stability of the mitotic spindle and midbody, which ensures appropriate chromosome segregation during mitotic division. The increase in acetyl-α-tubulin during YAP depletion may provide a lesion-halting mechanism in maintaining the microtubule structure. The depletion of YAP also results in multinuclearity and aneuploidy, which supports its role in stabilizing the mitotic spindle and midbody.


Asunto(s)
Neoplasias Pulmonares , Huso Acromático , Tubulina (Proteína) , Humanos , Línea Celular Tumoral , Células HeLa , Microtúbulos/metabolismo , Mitosis/genética , Huso Acromático/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Neoplasias Pulmonares/patología
9.
J Cell Sci ; 136(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37439191

RESUMEN

Abscission is the final stage of cytokinesis whereby the midbody, a thin intercellular bridge, is resolved to separate the daughter cells. Cytokinetic abscission is mediated by the endosomal sorting complex required for transport (ESCRT), a conserved membrane remodelling machinery. The midbody organiser CEP55 recruits early acting ESCRT factors such as ESCRT-I and ALIX (also known as PDCD6IP), which subsequently initiate the formation of ESCRT-III polymers that sever the midbody. We now identify UMAD1 as an ESCRT-I subunit that facilitates abscission. UMAD1 selectively associates with VPS37C and VPS37B, supporting the formation of cytokinesis-specific ESCRT-I assemblies. TSG101 recruits UMAD1 to the site of midbody abscission, to stabilise the CEP55-ESCRT-I interaction. We further demonstrate that the UMAD1-ESCRT-I interaction facilitates the final step of cytokinesis. Paradoxically, UMAD1 and ALIX co-depletion has synergistic effects on abscission, whereas ESCRT-III recruitment to the midbody is not inhibited. Importantly, we find that both UMAD1 and ALIX are required for the dynamic exchange of ESCRT-III subunits at the midbody. Therefore, UMAD1 reveals a key functional connection between ESCRT-I and ESCRT-III that is required for cytokinesis.


Asunto(s)
Citocinesis , Complejos de Clasificación Endosomal Requeridos para el Transporte , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Proteínas de Ciclo Celular
10.
Proteomics ; : e2300057, 2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507836

RESUMEN

Cell-derived extracellular vesicles (EVs) are evolutionary-conserved secretory organelles that, based on their molecular composition, are important intercellular signaling regulators. At least three classes of circulating EVs are known based on mechanism of biogenesis: exosomes (sEVs/Exos), microparticles (lEVs/MPs), and shed midbody remnants (lEVs/sMB-Rs). sEVs/Exos are of endosomal pathway origin, microparticles (lEVs/MPs) from plasma membrane blebbing and shed midbody remnants (lEVs/sMB-Rs) arise from symmetric cytokinetic abscission. Here, we isolate sEVs/Exos, lEVs/MPs, and lEVs/sMB-Rs secreted from human isogenic primary (SW480) and metastatic (SW620) colorectal cancer (CRC) cell lines in milligram quantities for label-free MS/MS-based proteomic profiling. Purified EVs revealed selective composition packaging of exosomal protein markers in SW480/SW620-sEVs/Exos, metabolic enzymes in SW480/SW620-lEVs/MPs, while centralspindlin complex proteins, nucleoproteins, splicing factors, RNA granule proteins, translation-initiation factors, and mitochondrial proteins selectively traffic to SW480/SW620- lEVs/sMB-Rs. Collectively, we identify 39 human cancer-associated genes in EVs; 17 associated with SW480-EVs, 22 with SW620-EVs. We highlight oncogenic receptors/transporters selectively enriched in sEVs/Exos (EGFR/FAS in SW480-sEVs/Exos and MET, TGFBR2, ABCB1 in SW620-sEVs/Exos). Interestingly, MDK, STAT1, and TGM2 are selectively enriched in SW480-lEVs/sMB-Rs, and ADAM15 to SW620-lEVs/sMB-Rs. Our study reveals sEVs/Exos, lEVs/MPs, and lEVs/sMB-Rs have distinct protein signatures that open potential diagnostic avenues of distinct types of EVs for clinical utility.

12.
Dev Cell ; 58(6): 474-488.e5, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36898376

RESUMEN

How canonical cytokinesis is altered during germ cell division to produce stable intercellular bridges, called "ring canals," is poorly understood. Here, using time-lapse imaging in Drosophila, we observe that ring canal formation occurs through extensive remodeling of the germ cell midbody, a structure classically associated with its function in recruiting abscission-regulating proteins in complete cytokinesis. Germ cell midbody cores reorganize and join the midbody ring rather than being discarded, and this transition is accompanied by changes in centralspindlin dynamics. The midbody-to-ring canal transformation is conserved in the Drosophila male and female germlines and during mouse and Hydra spermatogenesis. In Drosophila, ring canal formation depends on Citron kinase function to stabilize the midbody, similar to its role during somatic cell cytokinesis. Our results provide important insights into the broader functions of incomplete cytokinesis events across biological systems, such as those observed during development and disease states.


Asunto(s)
Citocinesis , Espermatogénesis , Masculino , Animales , Ratones , Citocinesis/fisiología , División Celular , Células Germinativas , Drosophila
13.
Adv Sci (Weinh) ; 10(12): e2204388, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36825683

RESUMEN

Chromatin bridges connecting the two segregating daughter nuclei arise from chromosome fusion or unresolved interchromosomal linkage. Persistent chromatin bridges are trapped in the cleavage plane, triggering cytokinesis delay. The trapped bridges occasionally break during cytokinesis, inducing DNA damage and chromosomal rearrangements. Recently, Caenorhabditis elegans LEM-3 and human TREX1 nucleases have been shown to process chromatin bridges. Here, it is shown that ANKLE1 endonuclease, the human ortholog of LEM-3, accumulates at the bulge-like structure of the midbody via its N-terminal ankyrin repeats. Importantly, ANKLE1-/- knockout cells display an elevated level of G1-specific 53BP1 nuclear bodies, prolonged activation of the DNA damage response, and replication stress. Increased DNA damage observed in ANKLE1-/- cells is rescued by inhibiting actin polymerization or reducing actomyosin contractility. ANKLE1 does not act in conjunction with structure-selective endonucleases, GEN1 and MUS81 in resolving recombination intermediates. Instead, ANKLE1 acts on chromatin bridges by priming TREX1 nucleolytic activity and cleaving bridge DNA to prevent the formation of micronuclei and cytosolic dsDNA that activate the cGAS-STING pathway. It is therefore proposed that ANKLE1 prevents DNA damage and autoimmunity by cleaving chromatin bridges to avoid catastrophic breakage mediated by actomyosin contractile forces.


Asunto(s)
Cromatina , Endonucleasas , Animales , Humanos , Endonucleasas/química , Endonucleasas/genética , Endonucleasas/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Núcleo Celular/metabolismo , ADN/metabolismo , Daño del ADN , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo
14.
Development ; 150(4)2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36789950

RESUMEN

We show that the zebrafish maternal-effect mutation too much information (tmi) corresponds to zebrafish prc1-like (prc1l), which encodes a member of the MAP65/Ase1/PRC1 family of microtubule-associated proteins. Embryos from tmi homozygous mutant mothers display cytokinesis defects in meiotic and mitotic divisions in the early embryo, indicating that Prc1l has a role in midbody formation during cell division at the egg-to-embryo transition. Unexpectedly, maternal Prc1l function is also essential for the reorganization of vegetal pole microtubules required for the segregation of dorsal determinants. Whereas Prc1 is widely regarded to crosslink microtubules in an antiparallel conformation, our studies provide evidence for an additional function of Prc1l in the bundling of parallel microtubules in the vegetal cortex of the early embryo during cortical rotation and prior to mitotic cycling. These findings highlight common yet distinct aspects of microtubule reorganization that occur during the egg-to-embryo transition, driven by maternal product for the midbody component Prc1l and required for embryonic cell division and pattern formation.


Asunto(s)
Citocinesis , Proteínas Asociadas a Microtúbulos , Pez Cebra , Animales , División Celular , Citocinesis/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo
15.
Cell Cycle ; 22(8): 951-966, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36691345

RESUMEN

The centrosome acts as a protein platform from which proteins are deployed to function throughout the cell cycle. Previously, we have shown that the prolyl isomerase Cyclophilin A (CypA) localizes to the centrosome in interphase and re-localizes to the midbody during mitosis where it functions in cytokinesis. In this study, investigation of CypA by SDS-PAGE during the cell cycle reveals that it undergoes a mobility shift during mitosis, indicative of a post-translational modification, which may correlate with its subcellular re-localization. Due to the lack of a phospho-specific antibody, we used site-directed mutagenesis to demonstrate that the previously identified serine 77 phosphorylation site within CypA is important for control of CypA centrosome localization. Furthermore, CypA is shown to interact with the mitotic NIMA-related kinase 2 (Nek2) during interphase and mitosis, while also interacting with the Nek2-antagonist PP1 during interphase but not during mitosis, suggesting a potential role for the Nek2-PP1 complex in CypA phospho-regulation. In support of this, Nek2 is capable of phosphorylating CypA in vitro. Overall, this work reveals that phosphorylation of CypA at serine 77 is important for its release from the centrosome during mitosis and may be regulated by the activity of Nek2 and PP1 during the cell cycle.


Asunto(s)
Ciclofilina A , Proteínas Serina-Treonina Quinasas , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Mitosis , Centrosoma/metabolismo , Serina/metabolismo , Proteínas de Ciclo Celular/metabolismo
16.
Musculoskelet Surg ; 107(4): 447-453, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35945416

RESUMEN

PURPOSE: The main aim of the study is to assess clinical and functional outcomes of arthroscopic outside-in repair of isolated radial tears of the midbody of lateral meniscus in professional athletes and to evaluate the return to the sport activity after surgery. METHODS: A retrospective data collection on professional athletes with isolated complete lesion of the midbody of lateral meniscus, treated with arthroscopic outside-in repair was carried out. Outcome measures included functional assessment, Limb Symmetry Index (LSI) and Hamstring Quadriceps Ratio (HQR) and Lysholm score collected before surgery and at 4-month follow-up. Data on return to sport practice and re-injury were also retrieved. RESULTS: Fourteen patients satisfied the selection criteria. Full return to professional sport activity (Tegner 10) was registered in the 86% of the cohort at 4 months after the surgery. Functional testing of the athletes showed a return of the LSI and HQR to the pre-surgical condition, demonstrating a full recovery of the functional ability and muscle strength. Similarly, clinical evaluation through Lysholm score showed an improvement, reaching an average of 97.7 points at 4 months follow-up. CONCLUSION: A good functional recovery and a high rate of return to play has been observed in a population of professional athletes, at 4 months after outside-in repair.


Asunto(s)
Artroscopía , Meniscos Tibiales , Humanos , Meniscos Tibiales/cirugía , Estudios Retrospectivos , Atletas , Evaluación de Resultado en la Atención de Salud
17.
Life (Basel) ; 14(1)2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38255661

RESUMEN

BACKGROUND AND AIM: Our research aims to find correlations between the brain imaging performed at term-corrected age and the atypical general movement (GM) patterns noticed during the same visit a-cramped-synchronized (CS) or poor repertoire (PR)-in formerly premature neonates to provide evidence for the structures involved in the modulation of GM patterns that could be injured and result in the appearance of these patterns and further deficits. MATERIALS AND METHODS: A total of 44 preterm neonates ((mean GA, 33.59 weeks (+2.43 weeks)) were examined in the follow-up program at Life Memorial Hospital Bucharest at term-equivalent age (TEA). The GM and ultrasound examinations were performed by trained and certified specialists. Three GM pattens were noted (normal, PR, or CS), and the measurements of the following cerebral structures were conducted via head ultrasounds: ventricular index, the short and long axes of the lateral ventricles, the midbody distance of the lateral ventricle, the diagonal of the caudate nucleus, the width of the basal ganglia, the width of the interhemispheric fissure, the sinocortical width, the length and thickness of the callosal body, the anteroposterior diameter of the pons, the diameter of the vermis, and the transverse diameters of the cerebellum and vermis. The ultrasound measurements were compared between the groups in order to find statistically significant correlations by using the FANOVA test (significance p < 0.05). RESULTS: The presence of the CS movement pattern was significantly associated with an increased ventricular index (mean 11.36 vs. 8.90; p = 0.032), increased midbody distance of the lateral ventricle-CS versus PR (8.31 vs. 3.73; p = 0.001); CS versus normal (8.31 vs. 3.34; p = 0.001), increased long and short axes of the lateral ventricles (p < 0.001), and decreased width of the basal ganglia-CS versus PR (11.07 vs. 15.69; p = 0.001); CS versus normal pattern (11.07 vs. 15.15; p = 0.0010). The PR movement pattern was significantly associated with an increased value of the sinocortical width when compared to the CS pattern (p < 0.001) and a decreased anteroposterior diameter of the pons when compared to both the CS (12.06 vs. 16.83; p = 0.001) and normal (12.06 vs. 16.78; p = 0.001) patterns. The same correlations were present when the subgroup of infants with a GA ≤ 32 weeks was analyzed. CONCLUSIONS: Our study demonstrated that there are correlations between atypical GM patterns (cramped-synchronized-CS and poor repertoire-PR) and abnormalities in the dimensions of the structures measured via ultrasound at the term-equivalent age. The correlations could provide information about the structures that are affected and could lead to a lack of modulation in the GM patterns.

18.
Cells ; 11(21)2022 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-36359734

RESUMEN

The midbody is an organelle that forms between the two daughter cells during cytokinesis. It co-ordinates the abscission of the nascent daughter cells and is composed of a multitude of proteins that are meticulously arranged into distinct temporal and spatial localization patterns. However, very little is known about the mechanisms that regulate the localization and function of midbody proteins. Here, we analyzed the temporal and spatial profiles of key midbody proteins during mitotic exit under normal conditions and after treatment with drugs that affect phosphorylation and proteasome-mediated degradation to decipher the impacts of post-translational modifications on midbody protein dynamics. Our results highlighted that midbody proteins show distinct spatio-temporal dynamics during mitotic exit and cytokinesis that depend on both ubiquitin-mediated proteasome degradation and phosphorylation/de-phosphorylation. They also identified two discrete classes of midbody proteins: 'transient' midbody proteins-including Anillin, Aurora B and PRC1-which rapidly accumulate at the midbody after anaphase onset and then slowly disappear, and 'stable' midbody proteins-including CIT-K, KIF14 and KIF23-which instead persist at the midbody throughout cytokinesis and also post abscission. These two classes of midbody proteins display distinct interaction networks with ubiquitylation factors, which could potentially explain their different dynamics and stability during cytokinesis.


Asunto(s)
Citocinesis , Humanos , Citocinesis/fisiología , Células HeLa , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas
19.
BMC Biol ; 20(1): 172, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35922843

RESUMEN

BACKGROUND: ATP-dependent chromatin remodeling complexes are multi-protein machines highly conserved across eukaryotic genomes. They control sliding and displacing of the nucleosomes, modulating histone-DNA interactions and making nucleosomal DNA more accessible to specific binding proteins during replication, transcription, and DNA repair, which are processes involved in cell division. The SRCAP and p400/Tip60 chromatin remodeling complexes in humans and the related Drosophila Tip60 complex belong to the evolutionary conserved INO80 family, whose main function is promoting the exchange of canonical histone H2A with the histone variant H2A in different eukaryotic species. Some subunits of these complexes were additionally shown to relocate to the mitotic apparatus and proposed to play direct roles in cell division in human cells. However, whether this phenomenon reflects a more general function of remodeling complex components and its evolutionary conservation remains unexplored. RESULTS: We have combined cell biology, reverse genetics, and biochemical approaches to study the subcellular distribution of a number of subunits belonging to the SRCAP and p400/Tip60 complexes and assess their involvement during cell division progression in HeLa cells. Interestingly, beyond their canonical chromatin localization, the subunits under investigation accumulate at different sites of the mitotic apparatus (centrosomes, spindle, and midbody), with their depletion yielding an array of aberrant outcomes of mitosis and cytokinesis, thus causing genomic instability. Importantly, this behavior was conserved by the Drosophila melanogaster orthologs tested, despite the evolutionary divergence between fly and humans has been estimated at approximately 780 million years ago. CONCLUSIONS: Overall, our results support the existence of evolutionarily conserved diverse roles of chromatin remodeling complexes, whereby subunits of the SRCAP and p400/Tip60 complexes relocate from the interphase chromatin to the mitotic apparatus, playing moonlighting functions required for proper execution of cell division.


Asunto(s)
Proteínas de Drosophila , Histonas , Animales , Cromatina/genética , Ensamble y Desensamble de Cromatina , ADN/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HeLa , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Humanos , Nucleosomas , Huso Acromático/metabolismo , Factores de Transcripción/metabolismo
20.
Front Cell Dev Biol ; 10: 894434, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774230

RESUMEN

Intercellular lumen formation is a crucial aspect of animal development and physiology that involves a complex interplay between the molecular and physical properties of the constituent cells. Embryos of the invasive freshwater mussel Dreissena rostriformis are ideal models for studying this process due to the large intercellular cavities that readily form during blastomere cleavage. Using this system, we show that recruitment of the transmembrane water channel protein aquaporin exclusively to the midbody of intercellular cytokinetic bridges is critical for lumenogenesis. The positioning of aquaporin-positive midbodies thereby influences the direction of cleavage cavity expansion. Notably, disrupting cytokinetic bridge microtubules impairs not only lumenogenesis but also cellular osmoregulation. Our findings reveal a simple mechanism that provides tight spatial and temporal control over the formation of luminal structures and likely plays an important role in water homeostasis during early cleavage stages of a freshwater invertebrate species.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...