1
|
Tong YY, Wang BZ, Zhang YJ, Jiang LL, Ding XF, Zhou J, Zuo DY, Chen J, Zhu J, Chen G. Motor neuron and pancreas homeobox 1 (MNX1) suppresses Triple Negative Breast Cancer (TNBC) cell phagocytosis by macrophage through CD24 signaling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167763. [PMID: 40037472 DOI: 10.1016/j.bbadis.2025.167763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/08/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer with limited therapeutic options. Motor neuron and pancreas homeobox 1 (MNX1) has been implicated in tumor progression, yet its roles in TNBC immune evasion remain unexplored. In the present study, we analyzed TCGA datasets and performed immunohistochemistry to evaluate MNX1 expression in TNBC tissues. DNA Affinity Purification and sequencing (DAP-seq) were used to identify MNX1 binding motifs. The regulatory relationship between MNX1 and CD24 was validated through dual luciferase reporter assays and expression manipulation in TNBC cell lines. Macrophage-mediated phagocytosis was assessed using in vitro co-culture systems and a humanized macrophage immune reconstruction mouse xenograft model. We found MNX1 expression was significantly elevated in TNBC tissues and correlated with poor patient prognosis. MNX1-knockdown significantly inhibited MDA-MB-468 cell proliferation in vitro and xenograft growth in vivo. In MDA-MB-231 cell, MNX1-overexpression promoted cell proliferation in vitro. We identified "TAATTA" as the MNX1 binding motif and demonstrated that MNX1 directly activates CD24 transcription. MNX1 knockdown in MDA-MB-468 cells enhanced macrophage phagocytosis, while its overexpression in MDA-MB-231 cells reduced phagocytosis. In the humanized mouse model, MNX1 downregulation increased macrophage infiltration and suppressed tumor growth. In summary, our findings reveal that MNX1 promotes TNBC immune evasion through transcriptional regulation of CD24. SIGNIFICANCE: We showed that CD24 is a novel target of transcription factor MNX1. MNX1-driven CD24-overexpression enables TNBC cells to evade from phagocytosis in both co-culturing TNBC cells with macrophage and in humanized macrophage immune reconstruction mouse xenograft model.
Collapse
Affiliation(s)
- Ying-Ying Tong
- Department of Lab Medicine, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Bo-Ze Wang
- Department of Lab Medicine, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Yi-Jia Zhang
- Department of Lab Medicine, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Ling-Ling Jiang
- Department of Lab Medicine, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China
| | - Xiao-Fei Ding
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China
| | - Jun Zhou
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China
| | - Dai-Ying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jie Chen
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China.
| | - Jin Zhu
- Department of Breast Surgeon, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, PR China.
| | - Guang Chen
- Department of Lab Medicine, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China; Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, PR China.
| |
Collapse
|
2
|
Bruno F, Naselli F, Brancato D, Volpes S, Cardinale PS, Saccone S, Federico C, Caradonna F. Effects of Pterostilbene on the Cell Division Cycle of a Neuroblastoma Cell Line. Nutrients 2024; 16:4152. [PMID: 39683545 DOI: 10.3390/nu16234152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background. The "Cell Cycle Hypothesis" suggests that the abnormal re-entry of neurons into the cell division cycle leads to neurodegeneration, a mechanism supported by in vitro studies on neuronal-like cells treated with the hyperphosphorylating agent forskolin. Pterostilbene, a bioavailable compound found in foods such as blueberries and grapes, may exert neuroprotective effects and could serve as a potential adjunct therapy for neurodegenerative diseases. Methods. In this study, we investigated the effects of pterostilbene on neuronal-like cells derived from the human neuroblastoma SK-N-BE cell line, where cell cycle reactivation was induced by forskolin treatment. We analyzed molecular endpoints associated with differentiated versus replicative cell states, specifically the following: (a) the expression of cyclin CCND1, (b) the Ki67 cell proliferation marker, (c) the AT8 nuclear tau epitope, and (d) genome-wide DNA methylation changes. Results. Our findings indicate that pterostilbene exerts distinct effects on the cell division cycle depending on the cellular state, with neuroprotective benefits observed in differentiated neuronal-like cells, but not in cells undergoing induced division. Additionally, pterostilbene alters DNA methylation patterns. Conclusion. These results suggest that pterostilbene may offer neuroprotective advantages for differentiated neuronal-like cells. However, further studies are required to confirm these effects in vivo by examining specific biomarkers in human populations consuming pterostilbene-containing foods.
Collapse
Affiliation(s)
- Francesca Bruno
- Department Biological, Geological, and Environmental Sciences, University of Catania, 95124 Catania, Italy
| | - Flores Naselli
- Department Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90133 Palermo, Italy
| | - Desiree Brancato
- Department Biological, Geological, and Environmental Sciences, University of Catania, 95124 Catania, Italy
| | - Sara Volpes
- Department Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90133 Palermo, Italy
| | - Paola Sofia Cardinale
- Department Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90133 Palermo, Italy
| | - Salvatore Saccone
- Department Biological, Geological, and Environmental Sciences, University of Catania, 95124 Catania, Italy
- CERNUT, Interdepartmental Research Center in Nutraceutics and Health Products, 95125 Catania, Italy
| | - Concetta Federico
- Department Biological, Geological, and Environmental Sciences, University of Catania, 95124 Catania, Italy
- CERNUT, Interdepartmental Research Center in Nutraceutics and Health Products, 95125 Catania, Italy
| | - Fabio Caradonna
- Department Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90133 Palermo, Italy
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| |
Collapse
|
3
|
Brancato D, Bruno F, Coniglio E, Sturiale V, Saccone S, Federico C. The Chromatin Organization Close to SNP rs12913832, Involved in Eye Color Variation, Is Evolutionary Conserved in Vertebrates. Int J Mol Sci 2024; 25:6602. [PMID: 38928306 PMCID: PMC11204186 DOI: 10.3390/ijms25126602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
The most significant genetic influence on eye color pigmentation is attributed to the intronic SNP rs12913832 in the HERC2 gene, which interacts with the promoter region of the contiguous OCA2 gene. This interaction, through the formation of a chromatin loop, modulates the transcriptional activity of OCA2, directly affecting eye color pigmentation. Recent advancements in technology have elucidated the precise spatial organization of the genome within the cell nucleus, with chromatin architecture playing a pivotal role in regulating various genome functions. In this study, we investigated the organization of the chromatin close to the HERC2/OCA2 locus in human lymphocyte nuclei using fluorescence in situ hybridization (FISH) and high-throughput chromosome conformation capture (Hi-C) data. The 3 Mb of genomic DNA that belonged to the chromosomal region 15q12-q13.1 revealed the presence of three contiguous chromatin loops, which exhibited a different level of compaction depending on the presence of the A or G allele in the SNP rs12913832. Moreover, the analysis of the genomic organization of the genes has demonstrated that this chromosomal region is evolutionarily highly conserved, as evidenced by the analysis of syntenic regions in species from other Vertebrate classes. Thus, the role of rs12913832 variant is relevant not only in determining the transcriptional activation of the OCA2 gene but also in the chromatin compaction of a larger region, underscoring the critical role of chromatin organization in the proper regulation of the involved genes. It is crucial to consider the broader implications of this finding, especially regarding the potential regulatory role of similar polymorphisms located within intronic regions, which do not influence the same gene by modulating the splicing process, but they regulate the expression of adjacent genes. Therefore, caution should be exercised when utilizing whole-exome sequencing for diagnostic purposes, as intron sequences may provide valuable gene regulation information on the region where they reside. Thus, future research efforts should also be directed towards gaining a deeper understanding of the precise mechanisms underlying the role and mode of action of intronic SNPs in chromatin loop organization and transcriptional regulation.
Collapse
Affiliation(s)
| | | | | | | | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy; (D.B.); (F.B.); (E.C.); (V.S.); (C.F.)
| | | |
Collapse
|
4
|
Waraky A, Östlund A, Nilsson T, Weichenhan D, Lutsik P, Bähr M, Hey J, Tunali G, Adamsson J, Jacobsson S, Morsy MHA, Li S, Fogelstrand L, Plass C, Palmqvist L. Aberrant MNX1 expression associated with t(7;12)(q36;p13) pediatric acute myeloid leukemia induces the disease through altering histone methylation. Haematologica 2024; 109:725-739. [PMID: 37317878 PMCID: PMC10905087 DOI: 10.3324/haematol.2022.282255] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/05/2023] [Indexed: 06/16/2023] Open
Abstract
Certain subtypes of acute myeloid leukemia (AML) in children have inferior outcome, such as AML with translocation t(7;12)(q36;p13) leading to an MNX1::ETV6 fusion along with high expression of MNX1. We have identified the transforming event in this AML and possible ways of treatment. Retroviral expression of MNX1 was able to induce AML in mice, with similar gene expression and pathway enrichment to t(7;12) AML patient data. Importantly, this leukemia was only induced in immune incompetent mice using fetal but not adult hematopoietic stem and progenitor cells. The restriction in transforming capacity to cells from fetal liver is in alignment with t(7;12)(q36;p13) AML being mostly seen in infants. Expression of MNX1 led to increased histone 3 lysine 4 mono-, di- and trimethylation, reduction in H3K27me3, accompanied with changes in genome-wide chromatin accessibility and genome expression, likely mediated through MNX1 interaction with the methionine cycle and methyltransferases. MNX1 expression increased DNA damage, depletion of the Lin-/Sca1+/c-Kit+ population and skewing toward the myeloid lineage. These effects, together with leukemia development, were prevented by pre-treatment with the S-adenosylmethionine analog Sinefungin. In conclusion, we have shown the importance of MNX1 in development of AML with t(7;12), supporting a rationale for targeting MNX1 and downstream pathways.
Collapse
Affiliation(s)
- Ahmed Waraky
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, and; Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg
| | - Anders Östlund
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg
| | - Tina Nilsson
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg
| | - Pavlo Lutsik
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg
| | - Marion Bähr
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg
| | - Joschka Hey
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg
| | - Gürcan Tunali
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg
| | - Jenni Adamsson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg
| | - Susanna Jacobsson
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg
| | | | - Susann Li
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg
| | - Linda Fogelstrand
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, and; Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg
| | - Lars Palmqvist
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, and; Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg.
| |
Collapse
|
5
|
Sturiale V, Bruno F, Brancato D, D’Amico AG, Maugeri G, D’Agata V, Saccone S, Federico C. Cell Cycle Reactivation, at the Start of Neurodegeneration, Induced by Forskolin and Aniline in Differentiated Neuroblastoma Cells. Int J Mol Sci 2023; 24:14373. [PMID: 37762676 PMCID: PMC10531780 DOI: 10.3390/ijms241814373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
A characteristic hallmark of Alzheimer's disease (AD) is the intracellular accumulation of hyperphosphorylated tau protein, a phenomenon that appears to have associations with oxidative stress, double-stranded DNA breakage, and the de-condensation of heterochromatin. Re-entry into the cell division cycle appears to be involved in the onset of this neurodegenerative process. Indeed, the cell cycle cannot proceed regularly in the differentiated neurons leading to cell death. Here, we induced cell cycle reactivation in neuronal-like cells, obtained by neuroblastoma cells treated with retinoic acid, by exposure to forskolin or aniline. These compounds determine tau hyperphosphorylation or oxidative stress, respectively, resulting in the appearance of features resembling the start of neuronal degeneration typical of AD, such as tau hyperphosphorylation and re-entry into the cell cycle. Indeed, we detected an increased transcriptional level of cyclins and the appearance of a high number of mitotic cells. We also observed a delay in the initiation of the cell cycle when forskolin was co-administered with pituitary adenylate cyclase-activating polypeptide (PACAP). This delay was not observed when PACAP was co-administered with aniline. Our data demonstrate the relevance of tau hyperphosphorylation in initiating an ectopic cell cycle in differentiated neuronal cells, a condition that can lead to neurodegeneration. Moreover, we highlight the utility of neuroblastoma cell lines as an in vitro cellular model to test the possible neuroprotective effects of natural molecules.
Collapse
Affiliation(s)
- Valentina Sturiale
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Francesca Bruno
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Desiree Brancato
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, 95124 Catania, Italy; (V.S.); (F.B.)
| |
Collapse
|
6
|
Li JK, Liu H, Zhang HW, Li J, Liang ZT. A Positive Feedback Loop of E2F4-Mediated Activation of MNX1 Regulates Tumour Progression in Colorectal Cancer. J Cancer 2023; 14:2739-2750. [PMID: 37779874 PMCID: PMC10539396 DOI: 10.7150/jca.86718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/19/2023] [Indexed: 10/03/2023] Open
Abstract
Purpose: Colorectal cancer (CRC) is the 3rd most prevalent malignant tumour globally. Although significant strides have been made in diagnosis and treatment, its prognosis at the moment remains unpromising. Therefore, there is an urgent and desperate need to identify novel biomarkers of CRC and evaluate its mechanism of tumourigenesis and development. Methods: JASPAR and RNAinter databases are used to analyze target genes associated with colorectal cancer. Western blotting, q-PCR and immunohistochemistry et, al. were used to detect the level of MNX1 in patients with colorectal cancer, and Chip-PCR was used to detect the targeted binding ability of E2F4 and MNX1. The cells and animal models overexpressed MNX1 and E2F4 were constructed by shRNA transfection. Results: Herein, MNX1 was highly expressed and linked to favourable overall survival curves in colorectal cancer. The functional assay revealed that MNX1 overexpression could promote proliferation, migration, and invasion of CRC cells. Based on the prediction of the JASPAR and RNAinter databases, the transcription factor, E2F4, was bound to the MNX1 promoter region. The Chromatin Immunoprecipitation (ChIP) assay verified the interactions between MNX1 and E2F4 in CRC. Additionally, we found that sh-E2F4 markedly downregulated the MNX1 levels and reduced CRC progression in vivo and in vitro, which reversed MNX1 overexpression. Conclusion: Therefore, our research discovered that E2F4-mediated abnormal MNX1 expression promotes CRC progression and could become a novel diagnostic or therapeutic target of CRC.
Collapse
Affiliation(s)
- Jia-Ke Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Hai Liu
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Hui-Wen Zhang
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jun Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Zhuo-Tao Liang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
7
|
Wu J, Yue C, Xu W, Li H, Zhu J, Li L. MNX1 facilitates the malignant progress of lung adenocarcinoma through transcriptionally upregulating CCDC34. Oncol Lett 2023; 26:325. [PMID: 37415626 PMCID: PMC10320431 DOI: 10.3892/ol.2023.13911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/29/2023] [Indexed: 07/08/2023] Open
Abstract
Lung adenocarcinoma (LUAD) represents the most prevalent subtype of lung cancer and typically has high incidence and fatality rates. Motor neuron and pancreas homeobox 1 (MNX1) and coiled-coil domain-containing 34 (CCDC34) serve as oncogenes in multiple types of cancer. However, their role in LUAD remains to be elucidated. In the present study, bioinformatics analysis and LUAD cell lines were adopted to examine the expression of MNX1 and CCDC34. The proliferation, migration and invasion abilities of A549 cells were determined using Cell Counting Kit-8, colony formation, wound-healing and Transwell assay, and flow cytometry was conducted to assess cell cycle distribution and apoptosis. The interaction between MNX1 and CCDC34 was verified by luciferase reporter and chromatin immunoprecipitation assays. In addition, an in vivo animal model of LUAD was established for validation. The results demonstrated that both MNX1 and CCDC34 were upregulated in LUAD cell lines. MNX1 knockdown significantly suppressed cell proliferation, migration and invasion, hindered cell cycle progression and promoted cell apoptosis in vitro and inhibited tumor growth in vivo. However, the antitumor effect of MNX1 knockdown was weakened by simultaneous CCDC34 overexpression in vitro. In terms of mechanism, MNX1 was demonstrated to directly bind to the CCDC34 promoter and transcriptionally activate CCDC34 expression. In conclusion, the present study highlighted a critical role of the MNX1/CCDC34 axis in regulating LUAD progression, providing novel therapeutic targets for LUAD treatment.
Collapse
Affiliation(s)
- Junhua Wu
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Chongmei Yue
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Weiguo Xu
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Hui Li
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Jing Zhu
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| | - Lin Li
- Respiratory and Critical Care Medicine, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
- School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
8
|
Ragusa D, Dijkhuis L, Pina C, Tosi S. Mechanisms associated with t(7;12) acute myeloid leukaemia: from genetics to potential treatment targets. Biosci Rep 2023; 43:BSR20220489. [PMID: 36622782 PMCID: PMC9894016 DOI: 10.1042/bsr20220489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/10/2023] Open
Abstract
Acute myeloid leukaemia (AML), typically a disease of elderly adults, affects 8 children per million each year, with the highest paediatric incidence in infants aged 0-2 of 18 per million. Recurrent cytogenetic abnormalities contribute to leukaemia pathogenesis and are an important determinant of leukaemia classification. The t(7;12)(q36;p13) translocation is a high-risk AML subtype exclusively associated with infants and represents the second most common abnormality in this age group. Mechanisms of t(7;12) leukaemogenesis remain poorly understood. The translocation relocates the entire MNX1 gene within the ETV6 locus, but a fusion transcript is present in only half of the patients and its significance is unclear. Instead, research has focused on ectopic MNX1 expression, a defining feature of t(7;12) leukaemia, which has nevertheless failed to produce transformation in conventional disease models. Recently, advances in genome editing technologies have made it possible to recreate the t(7;12) rearrangement at the chromosomal level. Together with recent studies of MNX1 involvement using murine in vivo, in vitro, and organoid-based leukaemia models, specific investigation on the biology of t(7;12) can provide new insights into this AML subtype. In this review, we provide a comprehensive up-to-date analysis of the biological features of t(7;12), and discuss recent advances in mechanistic understanding of the disease which may deliver much-needed therapeutic opportunities to a leukaemia of notoriously poor prognosis.
Collapse
Affiliation(s)
- Denise Ragusa
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| | - Liza Dijkhuis
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
| | - Cristina Pina
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| | - Sabrina Tosi
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| |
Collapse
|
9
|
Ragusa D, Tosi S, Sisu C. Pan-Cancer Analysis Identifies MNX1 and Associated Antisense Transcripts as Biomarkers for Cancer. Cells 2022; 11:cells11223577. [PMID: 36429006 PMCID: PMC9688723 DOI: 10.3390/cells11223577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
The identification of diagnostic and prognostic biomarkers is a major objective in improving clinical outcomes in cancer, which has been facilitated by the availability of high-throughput gene expression data. A growing interest in non-coding genomic regions has identified dysregulation of long non-coding RNAs (lncRNAs) in several malignancies, suggesting a potential use as biomarkers. In this study, we leveraged data from large-scale sequencing projects to uncover the expression patterns of the MNX1 gene and its associated lncRNAs MNX1-AS1 and MNX1-AS2 in solid tumours. Despite many reports describing MNX1 overexpression in several cancers, limited studies exist on MNX1-AS1 and MNX1-AS2 and their potential as biomarkers. By employing clustering methods to visualise multi-gene relationships, we identified a discriminative power of the three genes in distinguishing tumour vs. normal samples in several cancers of the gastrointestinal tract and reproductive systems, as well as in discerning oesophageal and testicular cancer histological subtypes. Notably, the expressions of MNX1 and its antisenses also correlated with clinical features and endpoints, uncovering previously unreported associations. This work highlights the advantages of using combinatory expression patterns of non-coding transcripts of differentially expressed genes as clinical evaluators and identifies MNX1, MNX1-AS1, and MNX1-AS2 expressions as robust candidate biomarkers for clinical applications.
Collapse
Affiliation(s)
- Denise Ragusa
- Leukaemia and Chromosome Research Laboratory, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Centre for Genome Engineering and Maintenance (CenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
| | - Sabrina Tosi
- Leukaemia and Chromosome Research Laboratory, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Centre for Genome Engineering and Maintenance (CenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
| | - Cristina Sisu
- Centre for Genome Engineering and Maintenance (CenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
- Correspondence:
| |
Collapse
|
10
|
Transcription Factor Hb9 Is Expressed in Glial Cell Lineages in the Developing Mouse Spinal Cord. eNeuro 2022; 9:ENEURO.0214-22.2022. [PMID: 36265906 PMCID: PMC9636997 DOI: 10.1523/eneuro.0214-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/14/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Hb9 (Mnx1) is a transcription factor described as a spinal cord motor neuron (MN)-specific marker and critical factor for the postmitotic specification of these cells. To date, expression of Hb9 in other cell types has not been reported. We performed a fate-mapping approach to examine distributions of Hb9-expressing cells and their progeny ("Hb9-lineage cells") within the embryonic and adult spinal cord of Hb9cre;Ai14 mice. We found that Hb9-lineage cells are distributed in a gradient of increasing abundance throughout the rostrocaudal spinal cord axis during embryonic and postnatal stages. Furthermore, although the majority of Hb9-lineage cells at cervical spinal cord levels are MNs, at more caudal levels, Hb9-lineage cells include small-diameter dorsal horn neurons, astrocytes, and oligodendrocytes. In the peripheral nervous system, we observed a similar phenomenon with more abundant Hb9-lineage Schwann cells in muscles of the lower body versus upper body muscles. We cultured spinal cord progenitors in vitro and found that gliogenesis was increased by treatment with the caudalizing factor FGF-8B, while glial tdTomato expression was increased by treatment with both FGF-8B and GDF-11. Together, these observations suggest that early and transient expression of Hb9 in spinal cord neural progenitors may be induced by caudalizing factors such as FGF and GDF signaling. Furthermore, our work raises the possibility that early Hb9 expression may influence the development of spinal cord macroglia and Schwann cells, especially at caudal regions. Together, these findings highlight the importance of using caution when designing experiments using Hb9cre mice to perform spinal cord MN-specific manipulations.
Collapse
|
11
|
Surgical management of Currarino syndrome in elderly patient with infected pre-sacral mass: Technical nuances and review of literature. Clin Neurol Neurosurg 2022; 222:107470. [DOI: 10.1016/j.clineuro.2022.107470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/08/2022] [Indexed: 11/24/2022]
|
12
|
Gaggi G, Di Credico A, Guarnieri S, Mariggiò MA, Di Baldassarre A, Ghinassi B. Human mesenchymal amniotic fluid stem cells reveal an unexpected neuronal potential differentiating into functional spinal motor neurons. Front Cell Dev Biol 2022; 10:936990. [PMID: 35938174 PMCID: PMC9354810 DOI: 10.3389/fcell.2022.936990] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/04/2022] [Indexed: 11/26/2022] Open
Abstract
Human amniotic fluids stem cells (hAFSCs) can be easily isolated from the amniotic fluid during routinely scheduled amniocentesis. Unlike hiPSCs or hESC, they are neither tumorigenic nor immunogenic and their use does not rise ethical or safety issues: for these reasons they may represent a good candidate for the regenerative medicine. hAFSCs are generally considered multipotent and committed towards the mesodermal lineages; however, they express many pluripotent markers and share some epigenetic features with hiPSCs. Hence, we hypothesized that hAFSCs may overcome their mesodermal commitment differentiating into to ectodermal lineages. Here we demonstrated that by the sequential exposure to specific factors, hAFSCs can give rise to spinal motor neurons (MNs), as evidenced by the gradual gene and protein upregulation of early and late MN markers (PAX6, ISL1, HB9, NF-L, vAChT). When co-cultured with myotubes, hAFSCs-derived MNs were able to create functional neuromuscular junctions that induced robust skeletal muscle contractions. These data demonstrated the hAFSCs are not restricted to mesodermal commitment and can generate functional MNs thus outlining an ethically acceptable strategy for the study and treatment of the neurodegenerative diseases.
Collapse
Affiliation(s)
- Giulia Gaggi
- Department of Medicine and Sciences of Aging, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Andrea Di Credico
- Department of Medicine and Sciences of Aging, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Simone Guarnieri
- Department of Neuroscience, Imaging and Clinical Sciences, Chieti, Italy
- Functional Biotechnologies Lab, Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Maria Addolorata Mariggiò
- Department of Neuroscience, Imaging and Clinical Sciences, Chieti, Italy
- Functional Biotechnologies Lab, Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Sciences of Aging, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), Chieti, Italy
- *Correspondence: Angela Di Baldassarre,
| | - Barbara Ghinassi
- Department of Medicine and Sciences of Aging, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), Chieti, Italy
| |
Collapse
|
13
|
Chromosomal Rearrangements and Altered Nuclear Organization: Recent Mechanistic Models in Cancer. Cancers (Basel) 2021; 13:cancers13225860. [PMID: 34831011 PMCID: PMC8616464 DOI: 10.3390/cancers13225860] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary New methodologies and technologies developed in the last few decades have highlighted the precise spatial organization of the genome into the cell nucleus, with chromatin architecture playing a central role in controlling several genome functions. Genes are expressed in a well-defined way and at a well-defined time during cell differentiation, and alterations in genome organization can lead to genetic diseases, such as cancers. Here we review how the genome is organized in the cell nucleus and the evidence of genome misorganization leading to cancer diseases. Abstract The last decade has seen significant progress in understanding how the genome is organized spatially within interphase nuclei. Recent analyses have confirmed earlier molecular cytogenetic studies on chromosome positioning within interphase nuclei and provided new information about the topologically associated domains (TADs). Examining the nuances of how genomes are organized within interphase nuclei will provide information fundamental to understanding gene regulation and expression in health and disease. Indeed, the radial spatial positioning of individual gene loci within nuclei has been associated with up- and down-regulation of specific genes, and disruption of normal genome organization within nuclei will result in compromised cellular health. In cancer cells, where reorganization of the nuclear architecture may occur in the presence of chromosomal rearrangements such as translocations, inversions, or deletions, gene repositioning can change their expression. To date, very few studies have focused on radial gene positioning and the correlation to gene expression in cancers. Further investigations would improve our understanding of the biological mechanisms at the basis of cancer and, in particular, in leukemia initiation and progression, especially in those cases where the molecular consequences of chromosomal rearrangements are still unclear. In this review, we summarize the main milestones in the field of genome organization in the nucleus and the alterations to this organization that can lead to cancer diseases.
Collapse
|
14
|
De Pasquale V, Scerra G, Scarcella M, D'Agostino M, Pavone LM. Competitive binding of extracellular accumulated heparan sulfate reduces lysosomal storage defects and triggers neuronal differentiation in a model of Mucopolysaccharidosis IIIB. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119113. [PMID: 34329663 DOI: 10.1016/j.bbamcr.2021.119113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022]
Abstract
Mucopolysaccharidoses (MPSs) are a group of inherited lysosomal storage disorders associated with the deficiency of lysosomal enzymes involved in glycosaminoglycan (GAG) degradation. The resulting cellular accumulation of GAGs is responsible for widespread tissue and organ dysfunctions. The MPS III, caused by mutations in the genes responsible for the degradation of heparan sulfate (HS), includes four subtypes (A, B, C, and D) that present significant neurological manifestations such as progressive cognitive decline and behavioral disorders. The established treatments for the MPS III do not cure the disease but only ameliorate non-neurological clinical symptoms. We previously demonstrated that the natural variant of the hepatocyte growth factor NK1 reduces the lysosomal pathology and reactivates impaired growth factor signaling in fibroblasts from MPS IIIB patients. Here, we show that the recombinant NK1 is effective in rescuing the morphological and functional dysfunctions of lysosomes in a neuronal cellular model of the MPS IIIB. More importantly, NK1 treatment is able to stimulate neuronal differentiation of neuroblastoma SK-NBE cells stable silenced for the NAGLU gene causative of the MPS IIIB. These results provide the basis for the development of a novel approach to possibly correct the neurological phenotypes of the MPS IIIB as well as of other MPSs characterized by the accumulation of HS and progressive neurodegeneration.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80127 Naples, Italy
| | - Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Melania Scarcella
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
15
|
High-Throughput Fluorescence-Based Screen Identifies the Neuronal MicroRNA miR-124 as a Positive Regulator of Alphavirus Infection. J Virol 2020; 94:JVI.02145-19. [PMID: 32102877 DOI: 10.1128/jvi.02145-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/16/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs which act by modulating the expression of target genes. In addition to their role in maintaining essential physiological functions in the cell, miRNAs can also regulate viral infections. They can do so directly by targeting RNAs of viral origin or indirectly by targeting host mRNAs, and this can result in a positive or negative outcome for the virus. Here, we performed a fluorescence-based miRNA genome-wide screen in order to identify cellular miRNAs involved in the regulation of arbovirus infection in human cells. We identified 16 miRNAs showing a positive effect on Sindbis virus (SINV) expressing green fluorescent protein (GFP), among which were a number of neuron-specific ones such as miR-124. We confirmed that overexpression of miR-124 increases both SINV structural protein translation and viral production and that this effect is mediated by its seed sequence. We further demonstrated that the SINV genome possesses a binding site for miR-124. Both inhibition of miR-124 and silent mutations to disrupt this binding site in the viral RNA abolished positive regulation. We also proved that miR-124 inhibition reduces SINV infection in human differentiated neuronal cells. Finally, we showed that the proviral effect of miR-124 is conserved in other alphaviruses, as its inhibition reduces chikungunya virus (CHIKV) production in human cells. Altogether, our work expands the panel of positive regulation of the viral cycle by direct binding of host miRNAs to the viral RNA and provides new insights into the role of cellular miRNAs as regulators of alphavirus infection.IMPORTANCE Arthropod-borne (arbo) viruses are part of a class of pathogens that are transmitted to their final hosts by insects. Because of climate change, the habitat of some of these insects, such as mosquitoes, is shifting, thereby facilitating the emergence of viral epidemics. Among the pathologies associated with arbovirus infection, neurological diseases such as meningitis and encephalitis represent a significant health burden. Using a genome-wide miRNA screen, we identified neuronal miR-124 as a positive regulator of the Sindbis and chikungunya alphaviruses. We also showed that this effect was in part direct, thereby opening novel avenues to treat alphavirus infections.
Collapse
|
16
|
Chen H, Zeng L, Zheng W, Li X, Lin B. Increased Expression of microRNA-141-3p Improves Necrotizing Enterocolitis of Neonates Through Targeting MNX1. Front Pediatr 2020; 8:385. [PMID: 32850524 PMCID: PMC7399201 DOI: 10.3389/fped.2020.00385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: MicroRNA-141-3p (miR-141-3p) has been investigated in various kinds of cancers. This research delves into the functions and regulatory mechanisms of miR-141-3p in necrotizing enterocolitis (NEC) of neonates. Methods: NEC tissues were obtained from neonatal mice, and subsequently, expression of miR-141-3p and motor neuron and pancreas homeobox 1 (MNX1) was assayed via RT-qPCR. Moreover, the intestinal histopathological changes and histiocytic apoptosis were observed via hematoxylin and eosin (H&E) and TUNEL staining. The correlative inflammatory factors and oxidative stress markers were evaluated to uncover the influence of miR-141-3p in NEC tissue damage. Further, the relation between MNX1 and miR-141-3p was predicated, and the functions of MNX1 in inflammatory response and cell growth of IEC-6 cells were investigated. Results: Downregulated miR-141-3p and upregulated MNX1 were discovered in NEC tissues. Moreover, miR-141-3p clearly alleviated inflammation response and oxidative stress damage in NEC, which was achieved through regulating inflammatory cytokines (IL-1β, IL-6, and TNF-α) and oxidative stress markers (MPO, MDA, and SOD) expression. MNX1 was forecasted as a target gene of miR-141-3p; meanwhile, MNX1 overexpression overturned the influence of miR-141-3p in the inflammatory response and cell growth process of IEC-6 cells. Conclusion: These explorations reveal that increased expression of miR-141-3p could improve the damage to intestinal tissues in NEC through targeting MNX1. The research might exhibit a neoteric therapeutic strategy for NEC.
Collapse
Affiliation(s)
- Hui Chen
- Department of Neonatology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Lichun Zeng
- Department of Neonatology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Wei Zheng
- Department of Neonatology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Xiaoli Li
- Department of Neonatology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Baixing Lin
- Department of Neonatology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| |
Collapse
|
17
|
Federico C, Owoka T, Ragusa D, Sturiale V, Caponnetto D, Leotta CG, Bruno F, Foster HA, Rigamonti S, Giudici G, Cazzaniga G, Bridger JM, Sisu C, Saccone S, Tosi S. Deletions of Chromosome 7q Affect Nuclear Organization and HLXB9Gene Expression in Hematological Disorders. Cancers (Basel) 2019; 11:cancers11040585. [PMID: 31027247 PMCID: PMC6521283 DOI: 10.3390/cancers11040585] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/19/2019] [Indexed: 12/22/2022] Open
Abstract
The radial spatial positioning of individual gene loci within interphase nuclei has been associated with up- and downregulation of their expression. In cancer, the genome organization may become disturbed due to chromosomal abnormalities, such as translocations or deletions, resulting in the repositioning of genes and alteration of gene expression with oncogenic consequences. In this study, we analyzed the nuclear repositioning of HLXB9 (also called MNX1), mapping at 7q36.3, in patients with hematological disorders carrying interstitial deletions of 7q of various extents, with a distal breakpoint in 7q36. We observed that HLXB9 remains at the nuclear periphery, or is repositioned towards the nuclear interior, depending upon the compositional properties of the chromosomal regions involved in the rearrangement. For instance, a proximal breakpoint leading the guanine-cytosine (GC)-poor band 7q21 near 7q36 would bring HLXB9 to the nuclear periphery, whereas breakpoints that join the GC-rich band 7q22 to 7q36 would bring HLXB9 to the nuclear interior. This nuclear repositioning is associated with transcriptional changes, with HLXB9 in the nuclear interior becoming upregulated. Here we report an in cis rearrangement, involving one single chromosome altering gene behavior. Furthermore, we propose a mechanistic model for chromatin reorganization that affects gene expression via the influences of new chromatin neighborhoods.
Collapse
Affiliation(s)
- Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Temitayo Owoka
- Genome Engineering and Maintenance Network, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane UB8 3PH, UK.
| | - Denise Ragusa
- Genome Engineering and Maintenance Network, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane UB8 3PH, UK.
| | - Valentina Sturiale
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Domenica Caponnetto
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Claudia Giovanna Leotta
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Francesca Bruno
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Helen A Foster
- Department of Biological and Environmental Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK.
- College of Health and Life Science, Brunel University London, Kingston Lane UB8 3PH, UK.
| | - Silvia Rigamonti
- Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP), Centro Ricerca Tettamanti, Pediatric Department, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Giovanni Giudici
- Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP), Centro Ricerca Tettamanti, Pediatric Department, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Giovanni Cazzaniga
- Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP), Centro Ricerca Tettamanti, Pediatric Department, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Joanna M Bridger
- Genome Engineering and Maintenance Network, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane UB8 3PH, UK.
| | - Cristina Sisu
- College of Health and Life Science, Brunel University London, Kingston Lane UB8 3PH, UK.
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, via Androne 81, 95124 Catania CT, Italy.
| | - Sabrina Tosi
- Genome Engineering and Maintenance Network, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane UB8 3PH, UK.
| |
Collapse
|
18
|
Tian T, Wang M, Zhu Y, Zhu W, Yang T, Li H, Lin S, Dai C, Deng Y, Song D, Li N, Zhai Z, Dai ZJ. Expression, Clinical Significance, and Functional Prediction of MNX1 in Breast Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:399-406. [PMID: 30368216 PMCID: PMC6205149 DOI: 10.1016/j.omtn.2018.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 09/16/2018] [Accepted: 09/23/2018] [Indexed: 12/24/2022]
Abstract
Motor neuron and pancreas homeobox 1 (MNX1) is a key developmental gene. Previous studies found that it was upregulated in several tumors, but its role in breast cancer (BC) remains unclear. In order to have a better understanding of this gene in BC, we examined the expression of MNX1 in BC tissues and normal breast tissues by qRT-PCR and by analyzing data from The Cancer Genome Atlas (TCGA) database. We also assessed the association of MNX1 expression with BC clinicopathological features and investigated the impact of MNX1 on BC survival. Potential molecular function of MNX1 was predicted through protein-protein interactions and functional enrichment. The results showed that the expression of MNX1 was significantly increased in BC tissues, especially in the HER2-positive subtype, and MNX1 expression was associated with several clinical characteristics, including menopause status, receptor status, subtypes, tumor size, lymph node metastasis, and race. In addition, patients with higher MNX1 expression had poorer survival. Enrichment analysis suggested that MNX1 is probably involved in biological processes and pathways related to nuclear division, cell cycle, and p53 signaling. In conclusion, our study suggests that MNX1 may act as a tumor promoter in BC. We hope these findings will draw more attention to MNX1 in future cancer studies.
Collapse
Affiliation(s)
- Tian Tian
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Meng Wang
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yuyao Zhu
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, The George Washington University Medical School, Washington, DC 20052, USA
| | - Tielin Yang
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hongtao Li
- Department of Breast, Head and Neck Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumchi 830000, China
| | - Shuai Lin
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Cong Dai
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yujiao Deng
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Dingli Song
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Na Li
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Zhen Zhai
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Zhi-Jun Dai
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
19
|
Crestini A, Vona R, Lo Giudice M, Sbriccoli M, Piscopo P, Borrelli A, Rivabene R, Ricceri L, Mancini A, Confaloni A. Differentiation-Dependent Effects of a New Recombinant Manganese Superoxide Dismutase on Human SK-N-BE Neuron-Like Cells. Neurochem Res 2018; 44:400-411. [PMID: 30471001 DOI: 10.1007/s11064-018-2686-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 09/24/2018] [Accepted: 11/16/2018] [Indexed: 11/29/2022]
Abstract
We have recently isolated a new isoform of recombinant manganese superoxide dismutase (rMnSOD) which provides a potent antitumor activity and strongly counteracts the occurrence of oxidative stress and tissue inflammation. This isoform, in addition to the enzymatic action common to all SODs, also shows special functional and structural properties, essentially due to the presence of a first leader peptide that allows the protein to enter easily into cells. Among endogenous antioxidants, SOD constitutes the first line of natural defence against pathological effects induced by an excess of free radicals. Here, we firstly describe the effects of our rMnSOD administration on the proliferant and malignant undifferentiated human neuroblastoma SK-N-BE cell line. Moreover, we also test the effects of rMnSOD in the all trans retinoic-differentiated SK-N-BE neuron-like cells, a quiescent "not malignant" model. While rMnSOD showed an antitumor activity on proliferating cells, a poor sensitivity to rMnSOD overload in retinoid-differentiated neuron-like cells was observed. However, in the latter case, in presence of experimental-induced oxidative stress, overcharge of rMnSOD enhanced the oxidant effects, through an increase of H2O2 due to low activity of both catalase and glutathione peroxidase. In conclusion, our data show that rMnSOD treatment exerts differential effects, which depend upon both cell differentiation and redox balance, addressing attention to the potential use of the recombinant enzyme on differentiated neurons. These facts ultimately pave the way for further preclinical studies aimed at evaluation of rMnSOD effects in models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Rosa Vona
- Biomarkers Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Rome, Italy
| | - Maria Lo Giudice
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Marco Sbriccoli
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Antonella Borrelli
- Molecular Biology and Viral Oncology Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Roberto Rivabene
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Laura Ricceri
- Centre for Behavioural Sciences and Mental Health, Italian National Institute of Health, Rome, Italy
| | - Aldo Mancini
- Leadhexa Biotechnologies Inc., San Francisco, CA, USA
| | - Annamaria Confaloni
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy.
| |
Collapse
|
20
|
Serratrice N, Fievet L, Albader F, Scavarda D, Dufour H, Fuentes S. Multiple neurosurgical treatments for different members of the same family with Currarino syndrome. Neurochirurgie 2018; 64:211-215. [PMID: 29731315 DOI: 10.1016/j.neuchi.2018.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/02/2017] [Accepted: 01/27/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Currarino's syndrome (CS) is an autosomal dominant disorder of embryonic development causing a rare malformating syndrome characterized by a triad of an anorectal malformations, presacral mass (most commonly an anterior sacral meningocele) and sacral bony defects. Mutations of the HLXB9 gene have been identified in most CS cases, but a precise genotype-phenotype correlation has not been described so far. Family screening is obligatory. The diagnosis is usually made during childhood and rarely in adulthood. In this context, imaging, and especially MRI plays a major role in the diagnosis of this syndrome. Surgical management is provided by pediatric surgeons or neurosurgeons. FAMILIAL CASE REPORT Here, we present a family case report with CS requiring different neurosurgical management. The son, a 3-year-old boy, developed a tethered spinal cord syndrome associated to a lipoma of the filum terminale, a sacro-coccygeal teratoma and an anal adhesion. A combined surgical approach permitted a good evolution on the urinary and digestive functions despite a persistent fecal incontinence. The 2-year-old daughter presented with a cyst of the thyreoglossal tract infected and fistulized to the skin. She was also followed for a very small lipoma of the filum terminale that required a neurosurgical approach. The father, 44-year-old, manifested functional digestive and urinary disorders caused by a giant anterior sacral meningocele. The ligation of the neck of the cyst and aspiration of the liquid inside in full through a posterior partial approach permit a complete collapse of the cyst with an instantly satisfactory clinical outcome. CONCLUSION In these cases, cooperation between pediatric surgeons and neurosurgeons was crucial. The follow-up of these patients should be done in a spina bifida clinic. A geneticist evaluation must be offered to the patient in the case of a CS as well as a clinical evaluation of the relatives (parents, siblings).
Collapse
Affiliation(s)
- N Serratrice
- Department of neurosurgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| | - L Fievet
- Department of pediatric surgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| | - F Albader
- Department of neurosurgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| | - D Scavarda
- Department of pediatric neurosurgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| | - H Dufour
- Department of neurosurgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| | - S Fuentes
- Department of neurosurgery, La Timone hospital, Assistance publique-Hôpitaux de Marseille, 13005 Marseille, France.
| |
Collapse
|
21
|
Federico C, Gil L, Bruno F, D'Amico AG, D'Agata V, Saccone S. Phosphorylated nucleolar Tau protein is related to the neuronal in vitro differentiation. Gene 2018; 664:1-11. [PMID: 29684490 DOI: 10.1016/j.gene.2018.04.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 02/02/2023]
Abstract
Tau is a multifunctional protein, originally identified as a cytoplasmic protein associated with microtubules. It is codified by the MAPT gene, and the alternative splicing, in the neuronal cells, results in six different isoforms. Tau was subsequently observed in the cell nucleus, where its function is not yet clearly understood. Here, we studied the MAPT gene and the cellular localization of the AT8 and Tau-1 epitopes of Tau protein, in the SK-N-BE cell line, which differentiates in neuronal-like cells after retinoic acid treatment. These epitopes correspond to the phosphorylated Ser202/Thr205 and unphosphorylated Pro189/Gly207 amino acid residues, respectively, possibly involved in conformational changes of the protein. Our results demonstrated the presence of the smaller Tau isoform (352 amino acids), whose amount increases in differentiated SK-N-BE cells, with Tau-1/AT8 nuclear distribution related to the differentiation process. Tau-1 showed a spot-like nucleolar localization, in both replicative and differentiated cells, while AT8 was only detected in the differentiated cells, diffusely occupying the entire nucleolar region. Moreover, in the replicative cells exposed to actinomycin-D, AT8 and Tau-1 move to the nucleolar periphery and colocalize, in few spots, with the upstream binding transcription factor (UBTF). Our results, also obtained with lymphocytes exposed to the mitogenic compound phytohaemagglutinin, indicate the AT8 epitope of Tau as a marker of neuronal cell differentiation, whose presence in the nucleolus appears to be related to rDNA transcriptional inactivation.
Collapse
Affiliation(s)
- Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | - Laura Gil
- Department of Genetics, Medical School, University "Alfonso X el Sabio", Madrid, Spain
| | - Francesca Bruno
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | - Agata Grazia D'Amico
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy; Department of Human Science and Promotion of Quality of Life, San Raffaele Open University of Rome, Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy.
| |
Collapse
|
22
|
Iarovaia OV, Ioudinkova ES, Razin SV, Vassetzky YS. Role of the Nucleolus in Rearrangements of the IGH Locus. Mol Biol 2018. [DOI: 10.1134/s0026893317050211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
23
|
Gassó P, Mas S, Rodríguez N, Boloc D, García-Cerro S, Bernardo M, Lafuente A, Parellada E. Microarray gene-expression study in fibroblast and lymphoblastoid cell lines from antipsychotic-naïve first-episode schizophrenia patients. J Psychiatr Res 2017; 95:91-101. [PMID: 28822801 DOI: 10.1016/j.jpsychires.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
Schizophrenia (SZ) is a chronic psychiatric disorder whose onset of symptoms occurs in late adolescence and early adulthood. The etiology is complex and involves important gene-environment interactions. Microarray gene-expression studies on SZ have identified alterations in several biological processes. The heterogeneity in the results can be attributed to the use of different sample types and other important confounding factors including age, illness chronicity and antipsychotic exposure. The aim of the present microarray study was to analyze, for the first time to our knowledge, differences in gene expression profiles in 18 fibroblast (FCLs) and 14 lymphoblastoid cell lines (LCLs) from antipsychotic-naïve first-episode schizophrenia (FES) patients and healthy controls. We used an analytical approach based on protein-protein interaction network construction and functional annotation analysis to identify the biological processes that are altered in SZ. Significant differences in the expression of 32 genes were found when LCLs were assessed. The network and gene set enrichment approach revealed the involvement of similar biological processes in FCLs and LCLs, including apoptosis and related biological terms such as cell cycle, autophagy, cytoskeleton organization and response to stress and stimulus. Metabolism and other processes, including signal transduction, kinase activity and phosphorylation, were also identified. These results were replicated in two independent cohorts using the same analytical approach. This provides more evidence for altered apoptotic processes in antipsychotic-naïve FES patients and other important biological functions such as cytoskeleton organization and metabolism. The convergent results obtained in both peripheral cell models support their usefulness for transcriptome studies on SZ.
Collapse
Affiliation(s)
- Patricia Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sergi Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | | | - Daniel Boloc
- Dept. of Basic Clinical Practice, University of Barcelona, Spain
| | | | - Miquel Bernardo
- Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Dept. of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Amalia Lafuente
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Eduard Parellada
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| |
Collapse
|
24
|
Gil L, Federico C, Pinedo F, Bruno F, Rebolledo AB, Montoya JJ, Olazabal IM, Ferrer I, Saccone S. Aging dependent effect of nuclear tau. Brain Res 2017; 1677:129-137. [PMID: 28974363 DOI: 10.1016/j.brainres.2017.09.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/11/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
Tau protein is characterized by a complex pattern of phosphorylation and is localized in the cytoplasm and nucleus in both neuronal and non-neuronal cells. Human AT100 nuclear tau, endowed by phosphorylation in Thr212/Ser214, was recently shown to decline in cornus ammonis 1 (CA1) and dentate gyrus (DG) in Alzheimer's disease (AD), but a defined function for this nuclear tau remains unclear. Here we show that AT100 progressively increases in the nuclei of neuronal and non-neuronal cells during aging, and decreases in the more severe AD stages, as recently shown, and in cancer cells (colorectal adenocarcinoma and breast cancer). AT100, in addition to a co-localization with the DAPI-positive heterochromatin, was detected in the nucleolus of pyramidal cells from the CA1 region, shown to be at its highest level in the more senescent cells and in the first stage of AD (ADI), and disappearing in the more severe AD cases (ADIV). Taking into account the nuclear distribution of AT100 during cell aging and its relation to the chromatin changes observed in degenerated neurons, as well as in cancerous cells, which are both cellular pathologies associated with age, we can consider the Thr212/Ser214 phosphorylated nuclear tau as a molecular marker of cell aging.
Collapse
Affiliation(s)
- Laura Gil
- Department of Genetics, Medical School, University "Alfonso X el Sabio", Madrid, Spain
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | - Fernando Pinedo
- Hospital Universitario Fundación Alcorcón, Department of Pathology, Alcorcon, Spain
| | - Francesca Bruno
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy
| | - Ana B Rebolledo
- Hospital Universitario Fundación Alcorcón, Department of Pathology, Alcorcon, Spain
| | - Juan J Montoya
- Department of Genetics, Medical School, University "Alfonso X el Sabio", Madrid, Spain
| | - Isabel M Olazabal
- Department of Genetics, Medical School, University "Alfonso X el Sabio", Madrid, Spain
| | - Isidre Ferrer
- Institut Neuropatologia - Hospital Universitari de Bellvitge, Barcelona, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Spain
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, Italy.
| |
Collapse
|
25
|
Bone morphogenic protein BMP7 induces adipocyte differentiation and uncoupling protein UCP1 expression in human bone marrow mesenchymal stem cells. RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2017. [DOI: 10.1007/s12210-017-0643-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
Genomic properties of chromosomal bands are linked to evolutionary rearrangements and new centromere formation in primates. Chromosome Res 2017; 25:261-276. [PMID: 28717965 DOI: 10.1007/s10577-017-9560-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 10/19/2022]
Abstract
Chromosomal rearrangements in humans are largely related to pathological conditions, and phenotypic effects are also linked to alterations in the expression profile following nuclear relocation of genes between functionally different compartments, generally occupying the periphery or the inner part of the cell nuclei. On the other hand, during evolution, chromosomal rearrangements may occur apparently without damaging phenotypic effects and are visible in currently phylogenetically related species. To increase our insight into chromosomal reorganisation in the cell nucleus, we analysed 18 chromosomal regions endowed with different genomic properties in cell lines derived from eight primate species covering the entire evolutionary tree. We show that homologous loci, in spite of their evolutionary relocation along the chromosomes, generally remain localised to the same functional compartment of the cell nuclei. We conclude that evolutionarily successful chromosomal rearrangements are those that leave the nuclear position of the regions involved unchanged. On the contrary, in pathological situations, the effect typically observed is on gene structure alteration or gene nuclear reposition. Moreover, our data indicate that new centromere formation could potentially occur everywhere in the chromosomes, but only those emerging in very GC-poor/gene-poor regions, generally located in the nuclear periphery, have a high probability of being retained through evolution. This suggests that, in the cell nucleus of related species, evolutionary chromosomal reshufflings or new centromere formation does not alter the functionality of the regions involved or the interactions between different loci, thus preserving the expression pattern of orthologous genes.
Collapse
|
27
|
Tawamie H, Martianov I, Wohlfahrt N, Buchert R, Mengus G, Uebe S, Janiri L, Hirsch FW, Schumacher J, Ferrazzi F, Sticht H, Reis A, Davidson I, Colombo R, Abou Jamra R. Hypomorphic Pathogenic Variants in TAF13 Are Associated with Autosomal-Recessive Intellectual Disability and Microcephaly. Am J Hum Genet 2017; 100:555-561. [PMID: 28257693 DOI: 10.1016/j.ajhg.2017.01.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/25/2017] [Indexed: 10/20/2022] Open
Abstract
In two independent consanguineous families each with two children affected by mild intellectual disability and microcephaly, we identified two homozygous missense variants (c.119T>A [p.Met40Lys] and c.92T>A [p.Leu31His]) in TATA-box-binding-protein-associated factor 13 (TAF13). Molecular modeling suggested a pathogenic effect of both variants through disruption of the interaction between TAF13 and TAF11. These two proteins form a histone-like heterodimer that is essential for their recruitment into the general RNA polymerase II transcription factor IID (TFIID) complex. Co-immunoprecipitation in HeLa cells transfected with plasmids encoding TAF11 and TAF13 revealed that both variants indeed impaired formation of the TAF13-TAF11 heterodimer, thus confirming the protein modeling analysis. To further understand the functional role of TAF13, we performed RNA sequencing of neuroblastoma cell lines upon TAF13 knockdown. The transcriptional profile showed significant deregulation of gene expression patterns with an emphasis on genes related to neuronal and skeletal functions and those containing E-box motives in their promoters. Here, we expand the spectrum of TAF-associated phenotypes and highlight the importance of TAF13 in neuronal functions.
Collapse
|
28
|
Tosi S, Mostafa Kamel Y, Owoka T, Federico C, Truong TH, Saccone S. Paediatric acute myeloid leukaemia with the t(7;12)(q36;p13) rearrangement: a review of the biological and clinical management aspects. Biomark Res 2015; 3:21. [PMID: 26605042 PMCID: PMC4657620 DOI: 10.1186/s40364-015-0041-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/25/2015] [Indexed: 12/05/2022] Open
Abstract
The presence of chromosomal abnormalities is one of the most important criteria for leukaemia diagnosis and management. Infant leukaemia is a rare disease that affects children in their first year of life. It has been estimated that approximately one third of infants with acute myeloid leukaemia harbour the t(7;12)(q36;p13) rearrangement in their leukaemic blasts. However, the WHO classification of acute myeloid leukaemia does not yet include the t(7;12) as a separate entity among the different genetic subtypes, although the presence of this chromosomal abnormality has been associated with an extremely poor clinical outcome. Currently, there is no consensus treatment for t(7;12) leukaemia patients. However, with the inferior outcome with the standard induction therapy, stem cell transplantation may offer a better chance for disease control. A better insight into the chromosome biology of this entity might shed some light into the pathogenic mechanisms arising from this chromosomal translocation, that at present are not fully understood. Further work is needed to improve our understanding of the molecular and genetic basis of this disorder. This will hopefully open some grounds for possible tailored treatment for this subset of very young patients with inferior disease outcome. This review aims at highlighting the cytogenetic features that characterise the t(7;12) leukaemias for a better detection of the abnormality in the diagnostic setting. We also review treatment and clinical outcome in the cases reported to date.
Collapse
Affiliation(s)
- Sabrina Tosi
- Leukaemia and Chromosome Research Laboratory, Division of Biosciences, Brunel University London, Middlesex, UB8 3PH UK
| | - Yasser Mostafa Kamel
- Leukaemia and Chromosome Research Laboratory, Division of Biosciences, Brunel University London, Middlesex, UB8 3PH UK
| | - Temitayo Owoka
- Leukaemia and Chromosome Research Laboratory, Division of Biosciences, Brunel University London, Middlesex, UB8 3PH UK
| | - Concetta Federico
- Dipartimento di Scienze Biologiche, Geologiche e Ambientali, Sezione di Biologia Animale, University of Catania, Catania, Italy
| | - Tony H Truong
- Division of Pediatric Oncology, Blood and Marrow Transplant, Alberta Children's Hospital, University of Calgary, Calgary, Canada
| | - Salvatore Saccone
- Dipartimento di Scienze Biologiche, Geologiche e Ambientali, Sezione di Biologia Animale, University of Catania, Catania, Italy
| |
Collapse
|
29
|
O'Brien LC, Keeney PM, Bennett JP. Differentiation of Human Neural Stem Cells into Motor Neurons Stimulates Mitochondrial Biogenesis and Decreases Glycolytic Flux. Stem Cells Dev 2015; 24:1984-94. [PMID: 25892363 DOI: 10.1089/scd.2015.0076] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Differentiation of human pluripotent stem cells (hPSCs) in vitro offers a way to study cell types that are not accessible in living patients. Previous research suggests that hPSCs generate ATP through anaerobic glycolysis, in contrast to mitochondrial oxidative phosphorylation (OXPHOS) in somatic cells; however, specialized cell types have not been assessed. To test if mitobiogenesis is increased during motor neuron differentiation, we differentiated human embryonic stem cell (hESC)- and induced pluripotent stem cell-derived human neural stem cells (hNSCs) into motor neurons. After 21 days of motor neuron differentiation, cells increased mRNA and protein levels of genes expressed by postmitotic spinal motor neurons. Electrophysiological analysis revealed voltage-gated currents characteristic of excitable cells and action potential formation. Quantitative PCR revealed an increase in peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), an upstream regulator of transcription factors involved in mitobiogenesis, and several of its downstream targets in hESC-derived cultures. This correlated with an increase in protein expression of respiratory subunits, but no increase in protein reflecting mitochondrial mass in either cell type. Respiration analysis revealed a decrease in glycolytic flux in both cell types on day 21 (D21), suggesting a switch from glycolysis to OXPHOS. Collectively, our findings suggest that mitochondrial biogenesis, but not mitochondrial mass, is increased during differentiation of hNSCs into motor neurons. These findings help us to understand human motor neuron mitobiogenesis, a process impaired in amyotrophic lateral sclerosis, a neurodegenerative disease characterized by death of motor neurons in the brain and spinal cord.
Collapse
Affiliation(s)
- Laura C O'Brien
- 1 Department of Physiology and Biophysics, Virginia Commonwealth University , Richmond, Virginia
| | - Paula M Keeney
- 2 VCU Parkinson's and Movement Disorders Center, Virginia Commonwealth University , Richmond, Virginia
| | - James P Bennett
- 1 Department of Physiology and Biophysics, Virginia Commonwealth University , Richmond, Virginia.,2 VCU Parkinson's and Movement Disorders Center, Virginia Commonwealth University , Richmond, Virginia.,3 Department of Neurology, Virginia Commonwealth University , Richmond, Virginia.,4 Department of Psychiatry, Virginia Commonwealth University , Richmond, Virginia
| |
Collapse
|
30
|
Gay S, Foiani M. Nuclear envelope and chromatin, lock and key of genome integrity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:267-330. [PMID: 26008788 DOI: 10.1016/bs.ircmb.2015.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
More than as an inert separation between the inside and outside of the nucleus, the nuclear envelope (NE) constitutes an active toll, which controls the import and export of molecules, and also a hub for a diversity of genomic processes, such as transcription, DNA repair, and chromatin dynamics. Proteins localized at the inner surface of the NE (such as lamins, nuclear pore proteins, lamin-associated proteins) interact with chromatin in a dynamic manner, contributing to the establishment of topological domains. In this review, we address the complex interplay between chromatin and NE. We discuss the divergence of this cross talk during evolution and comment both on the current established models and the most recent findings. In particular, we focus our attention on how the NE cooperates with chromatin in protecting the genome integrity.
Collapse
Affiliation(s)
- Sophie Gay
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Foiani
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy; Dipartimento di Scienze Biomolecolari e Biotecnologie, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|