1
|
Darieva Z, Zarrineh P, Phillips N, Mallen J, Garcia Mora A, Donaldson I, Bridoux L, Douglas M, Dias Henriques SF, Schulte D, Birket MJ, Bobola N. Ubiquitous MEIS transcription factors actuate lineage-specific transcription to establish cell fate. EMBO J 2025; 44:2232-2262. [PMID: 40021842 PMCID: PMC12000411 DOI: 10.1038/s44318-025-00385-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025] Open
Abstract
Control of gene expression is commonly mediated by distinct combinations of transcription factors (TFs). This cooperative action allows the integration of multiple biological signals at regulatory elements, resulting in highly specific gene expression patterns. It is unclear whether combinatorial binding is also necessary to bring together TFs with distinct biochemical functions, which collaborate to effectively recruit and activate RNA polymerase II. Using a cardiac differentiation model, we find that the largely ubiquitous homeodomain proteins MEIS act as actuators, fully activating transcriptional programs selected by lineage-restricted TFs. Combinatorial binding of MEIS with lineage-enriched TFs, GATA, and HOX, provides selectivity, guiding MEIS to function at cardiac-specific enhancers. In turn, MEIS TFs promote the accumulation of the methyltransferase KMT2D to initiate lineage-specific enhancer commissioning. MEIS combinatorial binding dynamics, dictated by the changing dosage of its partners, drive cells into progressive stages of differentiation. Our results uncover tissue-specific transcriptional activation as the result of ubiquitous actuator TFs harnessing general transcriptional activator at tissue-specific enhancers, to which they are directed by binding with lineage- and domain-specific TFs.
Collapse
Affiliation(s)
- Zoulfia Darieva
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Peyman Zarrineh
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Naomi Phillips
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Joshua Mallen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Araceli Garcia Mora
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ian Donaldson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Laure Bridoux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Megan Douglas
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Dorothea Schulte
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
| | - Matthew J Birket
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Nicoletta Bobola
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
De Palma FDE, Pol JG, Carbonnier V, Scuderi SA, Mannino D, Montégut L, Sauvat A, Perez-Lanzon M, Uribe-Carretero E, Guarracino M, Granata I, Calogero R, Del Monaco V, Montanaro D, Stoll G, Botti G, D'Aiuto M, Baldi A, D'Argenio V, Guigó R, Rezsohazy R, Kroemer G, Maiuri MC, Salvatore F. Epigenetic regulation of HOXA2 expression affects tumor progression and predicts breast cancer patient survival. Cell Death Differ 2025; 32:730-744. [PMID: 39833374 PMCID: PMC11982354 DOI: 10.1038/s41418-024-01430-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Accumulating evidence suggests that genetic and epigenetic biomarkers hold potential for enhancing the early detection and monitoring of breast cancer (BC). Epigenetic alterations of the Homeobox A2 (HOXA2) gene have recently garnered significant attention in the clinical management of various malignancies. However, the precise role of HOXA2 in breast tumorigenesis has remained elusive. To address this point, we conducted high-throughput RNA sequencing and DNA methylation array studies on laser-microdissected human BC samples, paired with normal tissue samples. Additionally, we performed comprehensive in silico analyses using large public datasets: TCGA and METABRIC. The diagnostic performance of HOXA2 was calculated by means of receiver operator characteristic curves. Its prognostic significance was assessed through immunohistochemical studies and Kaplan-Meier Plotter database interrogation. Moreover, we explored the function of HOXA2 and its role in breast carcinogenesis through in silico, in vitro, and in vivo investigations. Our work revealed significant hypermethylation and downregulation of HOXA2 in human BC tissues. Low HOXA2 expression correlated with increased BC aggressiveness and unfavorable patient survival outcomes. Suppression of HOXA2 expression significantly heightened cell proliferation, migration, and invasion in BC cells, and promoted tumor growth in mice. Conversely, transgenic HOXA2 overexpression suppressed these cellular processes and promoted apoptosis of cancer cells. Interestingly, a strategy of pharmacological demethylation successfully restored HOXA2 expression in malignant cells, reducing their neoplastic characteristics. Bioinformatics analyses, corroborated by in vitro experimentations, unveiled a novel implication of HOXA2 in the lipid metabolism of BC. Specifically, depletion of HOXA2 leaded to a concomitantly decreased expression of PPARγ and its target CIDEC, a master regulator of lipid droplet (LD) accumulation, thereby resulting in reduced LD abundance in BC cells. In summary, our study identifies HOXA2 as a novel prognosis-relevant tumor suppressor in the mammary gland.
Collapse
Affiliation(s)
- Fatima Domenica Elisa De Palma
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Jonathan G Pol
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Vincent Carbonnier
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sarah Adriana Scuderi
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Deborah Mannino
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Léa Montégut
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Allan Sauvat
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Perez-Lanzon
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisabet Uribe-Carretero
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Depto. Bioquimica y Biologia Molecular y Genetica, Facultad de Enfermeria y Terapia Ocupacional, Caceres, Spain
| | - Mario Guarracino
- University of Cassino and Southern Lazio, Cassino, Italy
- National Research University Higher School of Economics, Moscow, Russia
| | - Ilaria Granata
- National Research Council, Inst. for High-Performance Computing and Networking, Naples, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | | | - Gautier Stoll
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Gerardo Botti
- Department of Senology, Istituto Nazionale Tumori-IRCCS Fondazione Pascale, Naples, Italy
| | - Massimiliano D'Aiuto
- Department of Senology, Istituto Nazionale Tumori-IRCCS Fondazione Pascale, Naples, Italy
| | - Alfonso Baldi
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
| | - Roderic Guigó
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Bioinformatics and Genomics, Centre for Genomic Regulation (CRG), Barcelona, Catalonia, Spain
| | - René Rezsohazy
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Guido Kroemer
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Maria Chiara Maiuri
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Inter-University Center for multifactorial and multi genetic chronic human diseases, "Federico II"- Naples, Tor Vergata- Roma II and Chieti-Pescara Universities, Chieti-Pescara, Italy.
| |
Collapse
|
3
|
Yang RN, Li DZ, Liu A, Wei Y, Zhang RL, Abdelnabby H, Wang MQ. BarH1 regulates the expression of conserved odorant-binding protein 22 from Dastarcus helophoroides. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 178:104263. [PMID: 39880048 DOI: 10.1016/j.ibmb.2025.104263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Chemical signals are pivotal in establishing tritrophic interactions among host plants, herbivorous insects, and natural enemies. Previous studies have shown that evolutionarily conserved MaltOBPs in Monochamus alternatus and DhelOBPs in Dastarcus helophoroides contribute to the establishment of pine -pest - natural enemy tritrophic interactions by recognizing the same volatile emitted by the host during crucial developmental stages. We hypothesized that the transcriptional regulatory mechanisms of evolutionarily conserved OBPs respectively from pests and enemies are similar. In this study, we identified the promoter region of DhelOBP22 through chromosome walking and discovered that transcription factor BarH1, which have been proved to regulate the expression of MaltOBP19 in M. alternatus, regulates the expression of DhelOBP22 by binding to its promoter region, as evidenced by dual-luciferase assays and electrophoretic mobility shift assay (EMSA). When the BarH1 gene was silenced using RNAi, the expression of DhelOBP22 was inhibited, leading to the disappearance of the attracted behavior of mated female D. helophoroides adults towards camphene. This study underscores that conserved transcriptional regulation mechanisms play a role in regulating the expression of evolutionarily conserved OBPs respectively from herbivores and natural enemies.
Collapse
Affiliation(s)
- Rui-Nan Yang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; College of Bioscience and Resource Environment/Key Laboratory of Urban Agriculture (North China), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Beijing University of Agriculture, Beijing, 102206, China
| | - Dong-Zhen Li
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing, 100091, China
| | - Ao Liu
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yi Wei
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Rui-Lin Zhang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hazem Abdelnabby
- Department of Plant Protection, Faculty of Agriculture, Benha University, Banha, Qalyubia, 13736, Egypt
| | - Man-Qun Wang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
4
|
Marchese D, Evrard L, Bergiers I, Boas L, Duphénieux J, Hermant M, Pringels T, Zeqiri F, Pirson M, Twizere JC, Gofflot F, Rezsohazy R, Bridoux L. Homeodomain Involvement in Nuclear HOX Protein Homo- and Heterodimerization. Int J Mol Sci 2025; 26:423. [PMID: 39796276 PMCID: PMC11721573 DOI: 10.3390/ijms26010423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
HOX genes play essential roles in patterning the anteroposterior axis of animal embryos and in the formation of various organs. In mammals, there are 39 HOX genes organized into four clusters (HOXA-D) located on different chromosomes. In relationship with their orderly arrangement along the chromosomes, these genes show nested expression patterns which imply that embryonic territories co-express multiple HOX genes along the main body axis. Interactomic database entries, as well as a handful of publications, support that some HOX proteins can form homodimers or interact with other HOX proteins. However, the consequences of HOX protein interactions have been poorly investigated and remain largely elusive. In this study, we compiled a repository of all HOX-HOX interactions from available databases, and taking HOXA1, HOXA2, and HOXA5 as examples, we investigated the capacity of HOX proteins to form homo- and heterodimers. We revealed that while the DNA-binding domain, the homeodomain, is not necessary for HOXA1 homodimerization, the nuclear localization of the dimerization is dependent on the homeodomain, particularly the integrity of the third helix of HOXA1. Furthermore, we demonstrated that HOXA1 can influence the localization of HOXA1 when it is deprived of the homeodomain, increasing its abundance in the chromatin-containing fraction. Moreover, HOXA1 nuclear homodimerization occurs independently of the integrity of the hexapeptide and, consequently, of its well-known interactor, the homeodomain protein PBX. These results hint at a potential involvement of dimerization in the complex landscape of HOX regulatory mechanisms.
Collapse
Affiliation(s)
- Damien Marchese
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Laetitia Evrard
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Isabelle Bergiers
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Ludovic Boas
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Justine Duphénieux
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Maryse Hermant
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Tamara Pringels
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Fisnik Zeqiri
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Marc Pirson
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Jean-Claude Twizere
- Gembloux Agro-Bio Tech, University of Liège, Avenue de la Faculté de Gembloux, 5030 Gembloux, Belgium
| | - Françoise Gofflot
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - René Rezsohazy
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| | - Laure Bridoux
- Louvain Institute of Molecular Science and Technology, Université catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
5
|
Curt JR, Martín P, Foronda D, Hudry B, Kannan R, Shetty S, Merabet S, Saurin AJ, Graba Y, Sánchez- Herrero E. Ambivalent partnership of the Drosophila posterior class Hox protein Abdominal-B with Extradenticle and Homothorax. PLoS Genet 2025; 21:e1011355. [PMID: 39804927 PMCID: PMC11759358 DOI: 10.1371/journal.pgen.1011355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/24/2025] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Hox proteins, a sub-group of the homeodomain (HD) transcription factor family, provide positional information for axial patterning in development and evolution. Hox protein functional specificity is reached, at least in part, through interactions with Pbc (Extradenticle (Exd) in Drosophila) and Meis/Prep (Homothorax (Hth) in Drosophila) proteins. Most of our current knowledge of Hox protein specificity stems from the study of anterior and central Hox proteins, identifying the molecular and structural bases for Hox/Pbc/Meis-Prep cooperative action. Posterior Hox class proteins, Abdominal-B (Abd-B) in Drosophila and Hox9-13 in vertebrates, have been comparatively less studied. They strongly diverge from anterior and central class Hox proteins, with a low degree of HD sequence conservation and the absence of a core canonical Pbc interaction motif. Here we explore how Abd-B function interface with that of Exd/Hth using several developmental contexts, studying mutual expression control, functional dependency and intrinsic protein requirements. Results identify cross-regulatory interactions setting relative expression and activity levels required for proper development. They also reveal organ-specific requirement and a binary functional interplay with Exd and Hth, either antagonistic, as previously reported, or synergistic. This highlights context specific use of Exd/Hth, and a similar context specific use of Abd-B intrinsic protein requirements.
Collapse
Affiliation(s)
- Jesús R. Curt
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Paloma Martín
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - David Foronda
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Departamento de Medicina, Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, Madrid, Spain
| | - Bruno Hudry
- Aix-Marseille Univ., CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Parc Scientifique de Luminy, Marseille, France
- Institut de Biologie Valrose, Université Nice Sophia Antipolis, Faculté des Sciences Parc Valrose, Nice, France
| | - Ramakrishnan Kannan
- Molecular Genetics lab, Neurobiology Research Center (NRC), National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Srividya Shetty
- Molecular Genetics lab, Neurobiology Research Center (NRC), National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Samir Merabet
- Aix-Marseille Univ., CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Parc Scientifique de Luminy, Marseille, France
- Institut de Génétique Fonctionnelle, UMR 5242 CNRS/ENS Lyon, Lyon, France
| | - Andrew J. Saurin
- Aix-Marseille Univ., CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Parc Scientifique de Luminy, Marseille, France
| | - Yacine Graba
- Aix-Marseille Univ., CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Parc Scientifique de Luminy, Marseille, France
| | - Ernesto Sánchez- Herrero
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| |
Collapse
|
6
|
Frenette B, Guéno J, Houde N, Landry-Truchon K, Giguère A, Ashok T, Ryckman A, Morton BR, Mansfield JH, Jeannotte L. Loss of Hoxa5 function affects Hox gene expression in different biological contexts. Sci Rep 2024; 14:30903. [PMID: 39730789 DOI: 10.1038/s41598-024-81867-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Hoxa5 plays numerous roles in development, but its downstream molecular effects are mostly unknown. We applied bulk RNA-seq assays to characterize the transcriptional impact of the loss of Hoxa5 gene function in seven different biological contexts, including developing respiratory and musculoskeletal tissues that present phenotypes in Hoxa5 mouse mutants. This global analysis revealed few common transcriptional changes, suggesting that HOXA5 acts mainly via the regulation of context-specific effectors. However, Hox genes themselves appeared as potentially conserved targets of HOXA5 across tissues. Notably, a trend toward reduced expression of HoxA genes was observed in Hoxa5 null mutants in several tissue contexts. Comparative analysis of epigenetic marks along the HoxA cluster in lung tissue from two different Hoxa5 mutant mouse lines revealed limited effect of either mutation indicating that Hoxa5 gene targeting did not significantly perturb the chromatin landscape of the surrounding HoxA cluster. Combined with the shared impact of the two Hoxa5 mutant alleles on phenotype and Hox expression, these data argue against the contribution of local cis effects to Hoxa5 mutant phenotypes and support the notion that the HOXA5 protein acts in trans in the control of Hox gene expression.
Collapse
Affiliation(s)
- Béatrice Frenette
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Josselin Guéno
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Nicolas Houde
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada
| | - Kim Landry-Truchon
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada
| | - Anthony Giguère
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada
| | - Theyjasvi Ashok
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Abigail Ryckman
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Brian R Morton
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Jennifer H Mansfield
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA.
| | - Lucie Jeannotte
- Centre de Recherche sur le Cancer de L'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), 1401, 18e Rue, Québec, QC, G1J 1Z4, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada.
| |
Collapse
|
7
|
Cuvertino S, Garner T, Martirosian E, Walusimbi B, Kimber SJ, Banka S, Stevens A. Higher order interaction analysis quantifies coordination in the epigenome revealing novel biological relationships in Kabuki syndrome. Brief Bioinform 2024; 26:bbae667. [PMID: 39701600 DOI: 10.1093/bib/bbae667] [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: 07/25/2024] [Revised: 10/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Complex direct and indirect relationships between multiple variables, termed higher order interactions (HOIs), are characteristics of all natural systems. Traditional differential and network analyses fail to account for the omic datasets richness and miss HOIs. We investigated peripheral blood DNA methylation data from Kabuki syndrome type 1 (KS1) and control individuals, identified 2,002 differentially methylated points (DMPs), and inferred 17 differentially methylated regions, which represent only 189 DMPs. We applied hypergraph models to measure HOIs on all the CpGs and revealed differences in the coordination of DMPs with lower entropy and higher coordination of the peripheral epigenome in KS1 implying reduced network complexity. Hypergraphs also capture epigenomic trans-relationships, and identify biologically relevant pathways that escape the standard analyses. These findings construct the basis of a suitable model for the analysis of organization in the epigenome in rare diseases, which can be applied to investigate mechanism in big data.
Collapse
Affiliation(s)
- Sara Cuvertino
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Terence Garner
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Evgenii Martirosian
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Bridgious Walusimbi
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University Foundation NHS Trust Health Innovation Manchester, Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Siddharth Banka
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University Foundation NHS Trust Health Innovation Manchester, Manchester, UK
| | - Adam Stevens
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine, and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Khan MI, Bertram H, Schmitt AO, Ramzan F, Gültas M. Computational Identification of Milk Trait Regulation Through Transcription Factor Cooperation in Murciano-Granadina Goats. BIOLOGY 2024; 13:929. [PMID: 39596884 PMCID: PMC11591944 DOI: 10.3390/biology13110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
The Murciano-Granadina goat (MUG) is a renowned dairy breed, known for its adaptability and resilience, as well as for its exceptional milk traits characterized by high protein and fat content, along with low somatic cell counts. These traits are governed by complex biological processes, crucial in shaping phenotypic diversity. Thus, it is imperative to explore the factors regulating milk production and lactation for this breed. In this study, we investigated the genetic architecture of seven milk traits in MUGs, employing a two-step computational analysis to examine genotype-phenotype associations. Initially, a random forest algorithm identified the relative importance of each single-nucleotide polymorphism (SNP) in determining the traits of interest. The second step applied an information theory-based approach to exploring the complex genetic architecture of quantitative milk traits, focusing on epistatic interactions that may have been overlooked in the first step. These approaches allowed us to identify an almost distinct set of candidate genes for each trait. In contrast, by analyzing the promoter regions of these genes, we revealed common regulatory networks among the milk traits under study. These findings are crucial for understanding the molecular mechanisms underlying gene regulation, and they highlight the pivotal role of transcription factors (TFs) and their preferential interactions in the development of these traits. Notably, TFs such as DBP, HAND1E47, HOXA4, PPARA, and THAP1 were consistently identified for all traits, highlighting their important roles in immunity within the mammary gland and milk production during lactation.
Collapse
Affiliation(s)
- Muhammad Imran Khan
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany;
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany;
- Department of Livestock Production and Management, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi 46300, Pakistan
| | - Hendrik Bertram
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany;
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany;
| | - Armin Otto Schmitt
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany;
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| | - Faisal Ramzan
- Institute of Animal and Dairy Sciences, Faculty of Animal Husbandry, University of Agriculture, Faisalabad 38000, Pakistan;
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany;
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| |
Collapse
|
9
|
He X, Liao Y, Yu G, Wang S, Bao Y. Genome-wide association study reveals the underlying regulatory mechanisms of red blood traits in Anadara granosa. BMC Genomics 2024; 25:931. [PMID: 39367301 PMCID: PMC11452991 DOI: 10.1186/s12864-024-10857-3] [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: 05/16/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Anadara granosa, commonly known as the blood clam, exhibits the unusual characteristic of having red blood among invertebrates. There is significant individual variation in blood color intensity among blood clams; individuals with vibrant red blood are deemed healthier and exhibit stronger stress resistance. However, the molecular basis underlying these red blood traits (RBTs) remains poorly understood. RESULTS In this study, we performed genome-wide association studies (GWAS) in a population of 300 A. granosa individuals, focusing on RBTs as measured by hemoglobin concentration (HC), total hemocyte count (THC), and heme concentration (HEME). Our analysis identified 18 single nucleotide polymorphisms (SNPs) correlated with RBTs, subsequently selected 117 candidate genes within a 100 kb flanking region of these SNPs, potentially involved in the RBTs of A. granosa. Moreover, we discovered two haplotype blocks specifically associated with THC and HEME. Further analysis revealed eight genes (Septin7, Hox5, Cbfa2t3, Avpr1b, Hhex, Eif2ak3, Glrk, and Rpl35a) that significantly influence RBTs. Notably, a heterozygous A/T mutation in the 3'UTR of Cbfa2t3 was found to promote blood cell proliferation. These genes suggest that the hematopoietic function plays a significant role in the variability of RBTs in A. granosa. CONCLUSIONS Our findings reveal a conservation of the regulatory mechanisms of RBTs between blood clams and vertebrates. The results not only provide a scientific basis for selective breeding in blood clams, but also offer deeper insights into the evolutionary mechanisms of RBTs in invertebrates.
Collapse
Affiliation(s)
- Xin He
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai, 315604, China
- Key Laboratory of Aquatic Germplasm Resource of Zhejiang, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, China
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Yushan Liao
- Key Laboratory of Aquatic Germplasm Resource of Zhejiang, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, China
| | - Gaowei Yu
- Key Laboratory of Aquatic Germplasm Resource of Zhejiang, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, China
| | - Shi Wang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Yongbo Bao
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai, 315604, China.
- Key Laboratory of Aquatic Germplasm Resource of Zhejiang, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, China.
| |
Collapse
|
10
|
Kc R, López de Boer R, Lin M, Vagnozzi AN, Jeannotte L, Philippidou P. Multimodal Hox5 activity generates motor neuron diversity. Commun Biol 2024; 7:1166. [PMID: 39289460 PMCID: PMC11408534 DOI: 10.1038/s42003-024-06835-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Motor neurons (MNs) are the final output of circuits driving fundamental behaviors, such as respiration and locomotion. Hox proteins are essential in generating the MN diversity required for accomplishing these functions, but the transcriptional mechanisms that enable Hox paralogs to assign distinct MN subtype identities despite their promiscuous DNA binding motif are not well understood. Here we show that Hoxa5 modifies chromatin accessibility in all mouse spinal cervical MN subtypes and engages TALE co-factors to directly bind and regulate subtype-specific genes. We identify a paralog-specific interaction of Hoxa5 with the phrenic MN-specific transcription factor Scip and show that heterologous expression of Hoxa5 and Scip is sufficient to suppress limb-innervating MN identity. We also demonstrate that phrenic MN identity is stable after Hoxa5 downregulation and identify Klf proteins as potential regulators of phrenic MN maintenance. Our data identify multiple modes of Hoxa5 action that converge to induce and maintain MN identity.
Collapse
Affiliation(s)
- Ritesh Kc
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Raquel López de Boer
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Minshan Lin
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alicia N Vagnozzi
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Lucie Jeannotte
- Department of Molecular Biology, Medical Biochemistry & Pathology, Université Laval, Centre Recherche sur le Cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval (Oncology), Québec, Canada
| | - Polyxeni Philippidou
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
11
|
Yadav C, Yadav R, Nanda S, Ranga S, Ahuja P, Tanwar M. Role of HOX genes in cancer progression and their therapeutical aspects. Gene 2024; 919:148501. [PMID: 38670395 DOI: 10.1016/j.gene.2024.148501] [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: 01/19/2024] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
HOX genes constitute a family of evolutionarily conserved transcription factors that play pivotal roles in embryonic development, tissue patterning, and cell differentiation. These genes are essential for the precise spatial and temporal control of body axis formation in vertebrates. In addition to their developmental functions, HOX genes have garnered significant attention for their involvement in various diseases, including cancer. Deregulation of HOX gene expression has been observed in numerous malignancies, where they can influence tumorigenesis, progression, and therapeutic responses. This review provides an overview of the diverse roles of HOX genes in development, disease, and potential therapeutic targets, highlighting their significance in understanding biological processes and their potential clinical implications.
Collapse
Affiliation(s)
- Chetna Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Ritu Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana 124001, India.
| | - Smiti Nanda
- Retd. Senior Professor and Head, Department of Gynaecology and Obstetrics, Pt. B.D. Sharma University of Health Sciences, Rohtak 124001, India
| | - Shalu Ranga
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Parul Ahuja
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Mukesh Tanwar
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| |
Collapse
|
12
|
Rogoulenko E, Levy Y. Skipping events impose repeated binding attempts: profound kinetic implications of protein-DNA conformational changes. Nucleic Acids Res 2024; 52:6763-6776. [PMID: 38721783 PMCID: PMC11229352 DOI: 10.1093/nar/gkae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 07/09/2024] Open
Abstract
The kinetics of protein-DNA recognition, along with its thermodynamic properties, including affinity and specificity, play a central role in shaping biological function. Protein-DNA recognition kinetics are characterized by two key elements: the time taken to locate the target site amid various nonspecific alternatives; and the kinetics involved in the recognition process, which may necessitate overcoming an energetic barrier. In this study, we developed a coarse-grained (CG) model to investigate interactions between a transcription factor called the sex-determining region Y (SRY) protein and DNA, in order to probe how DNA conformational changes affect SRY-DNA recognition and binding kinetics. We find that, not only does a requirement for such a conformational DNA transition correspond to a higher energetic barrier for binding and therefore slower kinetics, it may further impede the recognition kinetics by increasing unsuccessful binding events (skipping events) where the protein partially binds its DNA target site but fails to form the specific protein-DNA complex. Such skipping events impose the need for additional cycles protein search of nonspecific DNA sites, thus significantly extending the overall recognition time. Our results highlight a trade-off between the speed with which the protein scans nonspecific DNA and the rate at which the protein recognizes its specific target site. Finally, we examine molecular approaches potentially adopted by natural systems to enhance protein-DNA recognition despite its intrinsically slow kinetics.
Collapse
Affiliation(s)
- Elena Rogoulenko
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yaakov Levy
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
13
|
Gawriyski L, Tan Z, Liu X, Chowdhury I, Malaymar Pinar D, Zhang Q, Weltner J, Jouhilahti EM, Wei GH, Kere J, Varjosalo M. Interaction network of human early embryonic transcription factors. EMBO Rep 2024; 25:1589-1622. [PMID: 38297188 PMCID: PMC10933267 DOI: 10.1038/s44319-024-00074-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/02/2024] Open
Abstract
Embryonic genome activation (EGA) occurs during preimplantation development and is characterized by the initiation of de novo transcription from the embryonic genome. Despite its importance, the regulation of EGA and the transcription factors involved in this process are poorly understood. Paired-like homeobox (PRDL) family proteins are implicated as potential transcriptional regulators of EGA, yet the PRDL-mediated gene regulatory networks remain uncharacterized. To investigate the function of PRDL proteins, we are identifying the molecular interactions and the functions of a subset family of the Eutherian Totipotent Cell Homeobox (ETCHbox) proteins, seven PRDL family proteins and six other transcription factors (TFs), all suggested to participate in transcriptional regulation during preimplantation. Using mass spectrometry-based interactomics methods, AP-MS and proximity-dependent biotin labeling, and chromatin immunoprecipitation sequencing we derive the comprehensive regulatory networks of these preimplantation TFs. By these interactomics tools we identify more than a thousand high-confidence interactions for the 21 studied bait proteins with more than 300 interacting proteins. We also establish that TPRX2, currently assigned as pseudogene, is a transcriptional activator.
Collapse
Affiliation(s)
- Lisa Gawriyski
- University of Helsinki, Institute of Biotechnology, Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Zenglai Tan
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Xiaonan Liu
- University of Helsinki, Institute of Biotechnology, Helsinki, Finland
| | | | - Dicle Malaymar Pinar
- University of Helsinki, Institute of Biotechnology, Helsinki, Finland
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Qin Zhang
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jere Weltner
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Eeva-Mari Jouhilahti
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Gong-Hong Wei
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Juha Kere
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Markku Varjosalo
- University of Helsinki, Institute of Biotechnology, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
14
|
Zhou Y, Wu Q, Guo Y. Deciphering the emerging landscape of HOX genes in cardiovascular biology, atherosclerosis and beyond (Review). Int J Mol Med 2024; 53:17. [PMID: 38131178 PMCID: PMC10781420 DOI: 10.3892/ijmm.2023.5341] [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: 08/05/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Atherosclerosis, a dominant driving force underlying multiple cardiovascular events, is an intertwined and chronic inflammatory disease characterized by lipid deposition in the arterial wall, which leads to diverse cardiovascular problems. Despite unprecedented advances in understanding the pathogenesis of atherosclerosis and the substantial decline in cardiovascular mortality, atherosclerotic cardiovascular disease remains a global public health issue. Understanding the molecular landscape of atherosclerosis is imperative in the field of molecular cardiology. Recently, compelling evidence has shown that an important family of homeobox (HOX) genes endows causality in orchestrating the interplay between various cardiovascular biological processes and atherosclerosis. Despite seemingly scratching the surface, such insight into the realization of biology promises to yield extraordinary breakthroughs in ameliorating atherosclerosis. Primarily recapitulated herein are the contributions of HOX in atherosclerosis, including diverse cardiovascular biology, knowledge gaps, remaining challenges and future directions. A snapshot of other cardiovascular biological processes was also provided, including cardiac/vascular development, cardiomyocyte pyroptosis/apoptosis, cardiac fibroblast proliferation and cardiac hypertrophy, which are responsible for cardiovascular disorders. Further in‑depth investigation of HOX promises to provide a potential yet challenging landscape, albeit largely undetermined to date, for partially pinpointing the molecular mechanisms of atherosclerosis. A plethora of new targeted therapies may ultimately emerge against atherosclerosis, which is rapidly underway. However, translational undertakings are crucially important but increasingly challenging and remain an ongoing and monumental conundrum in the field.
Collapse
Affiliation(s)
- Yu Zhou
- Medical College, Guizhou University, Guiyang, Guizhou 550025, P.R. China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yingchu Guo
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
15
|
Hakami MA, Hazazi A, Abdulaziz O, Almasoudi HH, Alhazmi AYM, Alkhalil SS, Alharthi NS, Alhuthali HM, Almalki WH, Gupta G, Khan FR. HOTAIR: A key regulator of the Wnt/β-catenin signaling cascade in cancer progression and treatment. Pathol Res Pract 2024; 253:154957. [PMID: 38000201 DOI: 10.1016/j.prp.2023.154957] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
The long non-coding RNA (lncRNA) HOTAIR occupies a central position in the complex domain of cancer biology, particularly concerning its intricate interplay with the Wnt/β-catenin signaling pathway. This comprehensive review explores the multifaceted interactions between HOTAIR and the Wnt/β-catenin cascade, elucidating their profound function in cancer growth, progression, and therapeutic strategies. The study commences by underscoring the pivotal role of the Wnt/β-catenin cascade in governing essential cellular activities, emphasizing its dysregulation as a linchpin in cancer initiation and advancement. It introduces HOTAIR as a crucial regulatory entity, influencing gene expression in both healthy and diseased. The core of this review plunges into the intricacies of HOTAIR's engagement with Wnt/β-catenin signaling. It unravels how HOTAIR, through epigenetic modifications and transcriptional control, exerts its influence over key pathway constituents, including β-catenin, Wnt ligands, and target genes. This influence drives unchecked cancer cell growth, invasion, and metastasis. Furthermore, the review underscores the clinical significance of the HOTAIR-Wnt/β-catenin interplay, elucidating its associations with diverse cancer subtypes, patient prognoses, and prospects as a therapy. It provides insights into ongoing research endeavors to develop HOTAIR-targeted treatments and initiatives to facilitate aberrant Wnt/β-catenin activation. Concluding on a forward-looking note, the article accentuates the broader implications of HOTAIR's involvement in cancer biology, including its contributions to therapy resistance and metastatic dissemination. It underscores the importance of delving deeper into these intricate molecular relationships to pave the way for groundbreaking cancer treatment.
Collapse
Affiliation(s)
- Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif Province, Saudi Arabia
| | - Hassan Hussain Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | | | - Samia S Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Nahed S Alharthi
- Department of Medical Laboratory Sciences. College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudia Arabia
| | - Hayaa M Alhuthali
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif Province, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia.
| |
Collapse
|
16
|
Bobola N, Sagerström CG. TALE transcription factors: Cofactors no more. Semin Cell Dev Biol 2024; 152-153:76-84. [PMID: 36509674 DOI: 10.1016/j.semcdb.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Exd/PBX, Hth/MEIS and PREP proteins belong to the TALE (three-amino-acid loop extension) superclass of transcription factors (TFs) with an atypical homedomain (HD). Originally discovered as "cofactors" to HOX proteins, revisiting their traditional role in light of genome-wide experiments reveals a strong and reproducible pattern of HOX and TALE co-occupancy across diverse embryonic tissues. While confirming that TALE increases HOX specificity and selectivity in vivo, this wider outlook also reveals novel aspects of HOX:TALE collaboration, namely that HOX TFs generally require pre-bound TALE factors to access their functional binding sites in vivo. In contrast to the restricted expression domains of HOX TFs, TALE factors are largely ubiquitous, and PBX and PREP are expressed at the earliest developmental stages. PBX and MEIS control development of many organs and tissues and their dysregulation is associated with congenital disease and cancer. Accordingly, many instances of TALE cooperation with non HOX TFs have been documented in various systems. The model that emerges from these studies is that TALE TFs create a permissive chromatin platform that is selected by tissue-restricted TFs for binding. In turn, HOX and other tissue-restricted TFs selectively convert a ubiquitous pool of low affinity TALE binding events into high confidence, tissue-restricted binding events associated with transcriptional activation. As a result, TALE:TF complexes are associated with active chromatin and domain/lineage-specific gene activity. TALE ubiquitous expression and broad genomic occupancy, as well as the increasing examples of TALE tissue-specific partners, reveal a universal and obligatory role for TALE in the control of tissue and lineage-specific transcriptional programs, beyond their initial discovery as HOX co-factors.
Collapse
Affiliation(s)
- Nicoletta Bobola
- School of Medical Sciences, University of Manchester, Manchester, UK.
| | - Charles G Sagerström
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Medical School, Aurora, CO, USA.
| |
Collapse
|
17
|
Li M, Bai G, Cen Y, Xie Q, Chen J, Chen J, Chen Q, Zhong W, Zhou X. Silencing HOXC13 exerts anti-prostate cancer effects by inducing DNA damage and activating cGAS/STING/IRF3 pathway. J Transl Med 2023; 21:884. [PMID: 38057852 PMCID: PMC10701956 DOI: 10.1186/s12967-023-04743-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Advanced prostate cancer (PCa) will develop into castration-resistant prostate cancer (CRPC) and lead to poor prognosis. As the primary subtype of CRPC, CRPC-AR accounts for the major induction of PCa heterogeneity. CRPC-AR is mainly driven by 25 transcription factors (TFs), which we speculate may be the key factors driving PCa toward CRPC. Therefore, it is necessary to clarify the key regulator and its molecular mechanism mediating PCa progression. METHODS Firstly, we downloaded transcriptomic data and clinical information from TCGA-PRAD. The characteristic gene cluster was identified by PPI clustering, GO enrichment, co-expression correlation and clinical feature analyses for 25 TFs. Then, the effects of 25 TFs expression on prognosis of PCa patients was analyzed using univariate Cox regression, and the target gene was identified. The expression properties of the target gene in PCa tissues were verified using tissue microarray. Meanwhile, the related mechanistic pathway of the target gene was mined based on its function. Next, the target gene was silenced by small interfering RNAs (siRNAs) for cellular function and mechanistic pathway validation. Finally, CIBERSORT algorithm was used to analyze the infiltration levels of 22 immune cells in PCa patients with low and high expression of target gene, and validated by assaying the expression of related immunomodulatory factor. RESULTS We found that HOX family existed independently in 25 TFs, among which HOXC10, HOXC12 and HOXC13 had unique clinical features and the PCa patients with high HOXC13 expression had the worst prognosis. In addition, HOXC13 was highly expressed in tumor tissues and correlated with Gleason score and pathological grade. In vitro experiments demonstrated that silencing HOXC13 inhibited 22RV1 and DU145 cell function by inducing cellular DNA damage and activating cGAS/STING/IRF3 pathway. Immune infiltration analysis revealed that high HOXC13 expression suppressed infiltration of γδ T cells and plasma cells and recruited M2 macrophages. Consistent with these results, silencing HOXC13 up-regulated the transcriptional expression of IFN-β, CCL2, CCL5 and CXCL10. CONCLUSION HOXC13 regulates PCa progression by mediating the DNA damage-induced cGAS/STING/IRF3 pathway and remodels TIME through regulation of the transcription of the immune factors IFN-β, CCL2, CCL5 and CXCL10.
Collapse
Affiliation(s)
- Maozhang Li
- School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Guangwei Bai
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Yi Cen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Qitong Xie
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Jiahong Chen
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Jia Chen
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Qingbiao Chen
- Department of Urology, The Second People's Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, 528000, China
| | - Weide Zhong
- School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Xiaobo Zhou
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China.
| |
Collapse
|
18
|
Heinze SD, Berger S, Engleitner S, Daube M, Hajnal A. Prolonging somatic cell proliferation through constitutive hox gene expression in C. elegans. Nat Commun 2023; 14:6850. [PMID: 37891160 PMCID: PMC10611754 DOI: 10.1038/s41467-023-42644-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
hox genes encode a conserved family of homeodomain transcription factors that are essential to determine the identity of body segments during embryogenesis and maintain adult somatic stem cells competent to regenerate organs. In contrast to higher organisms, somatic cells in C. elegans irreversibly exit the cell cycle after completing their cell lineage and the adult soma cannot regenerate. Here, we show that hox gene expression levels in C. elegans determine the temporal competence of somatic cells to proliferate. Down-regulation of the central hox gene lin-39 in dividing vulval cells results in their premature cell cycle exit, whereas constitutive lin-39 expression causes precocious Pn.p cell and sex myoblast divisions and prolongs the proliferative phase of the vulval cells past their normal point of arrest. Furthermore, ectopic expression of hox genes in the quiescent anchor cell re-activates the cell cycle and induces proliferation until young adulthood. Thus, constitutive expression of a single hox transcription factor is sufficient to prolong somatic cell proliferation beyond the restriction imposed by the cell lineage. The down-regulation of hox gene expression in most somatic cells at the end of larval development may be one cause for the absence of cell proliferation in adult C. elegans.
Collapse
Affiliation(s)
- Svenia D Heinze
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, CH-8057, Zürich, Switzerland
| | - Simon Berger
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Institute for Chemical- and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093, Zürich, Switzerland
| | - Stefanie Engleitner
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Molecular Life Science PhD Program, University and ETH Zürich, CH-8057, Zürich, Switzerland
| | - Michael Daube
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Alex Hajnal
- Department of Molecular Life Sciences, University Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
19
|
Hodkinson LJ, Smith C, Comstra HS, Ajani BA, Albanese EH, Arsalan K, Daisson AP, Forrest KB, Fox EH, Guerette MR, Khan S, Koenig MP, Lam S, Lewandowski AS, Mahoney LJ, Manai N, Miglay J, Miller BA, Milloway O, Ngo N, Ngo VD, Oey NF, Punjani TA, SiMa H, Zeng H, Schmidt CA, Rieder LE. A bioinformatics screen reveals hox and chromatin remodeling factors at the Drosophila histone locus. BMC Genom Data 2023; 24:54. [PMID: 37735352 PMCID: PMC10515271 DOI: 10.1186/s12863-023-01147-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/07/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Cells orchestrate histone biogenesis with strict temporal and quantitative control. To efficiently regulate histone biogenesis, the repetitive Drosophila melanogaster replication-dependent histone genes are arrayed and clustered at a single locus. Regulatory factors concentrate in a nuclear body known as the histone locus body (HLB), which forms around the locus. Historically, HLB factors are largely discovered by chance, and few are known to interact directly with DNA. It is therefore unclear how the histone genes are specifically targeted for unique and coordinated regulation. RESULTS To expand the list of known HLB factors, we performed a candidate-based screen by mapping 30 publicly available ChIP datasets of 27 unique factors to the Drosophila histone gene array. We identified novel transcription factor candidates, including the Drosophila Hox proteins Ultrabithorax (Ubx), Abdominal-A (Abd-A), and Abdominal-B (Abd-B), suggesting a new pathway for these factors in influencing body plan morphogenesis. Additionally, we identified six other factors that target the histone gene array: JIL-1, hormone-like receptor 78 (Hr78), the long isoform of female sterile homeotic (1) (fs(1)h) as well as the general transcription factors TBP associated factor 1 (TAF-1), Transcription Factor IIB (TFIIB), and Transcription Factor IIF (TFIIF). CONCLUSIONS Our foundational screen provides several candidates for future studies into factors that may influence histone biogenesis. Further, our study emphasizes the powerful reservoir of publicly available datasets, which can be mined as a primary screening technique.
Collapse
Affiliation(s)
- Lauren J Hodkinson
- Genetics and Molecular Biology graduate program, Emory University, Atlanta, GA, 30322, USA
| | - Connor Smith
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - H Skye Comstra
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Bukola A Ajani
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Eric H Albanese
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Kawsar Arsalan
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Alvaro Perez Daisson
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Katherine B Forrest
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Elijah H Fox
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Matthew R Guerette
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Samia Khan
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Madeleine P Koenig
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Shivani Lam
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Ava S Lewandowski
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Lauren J Mahoney
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Nasserallah Manai
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - JonCarlo Miglay
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Blake A Miller
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Olivia Milloway
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Nhi Ngo
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Vu D Ngo
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Nicole F Oey
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Tanya A Punjani
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - HaoMin SiMa
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Hollis Zeng
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Casey A Schmidt
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA.
| | - Leila E Rieder
- Department of Biology, Emory University, 1510 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Zhang Z, Peng J, Li B, Wang Z, Wang H, Wang Y, Hong L. HOXA1 promotes aerobic glycolysis and cancer progression in cervical cancer. Cell Signal 2023; 109:110747. [PMID: 37286120 DOI: 10.1016/j.cellsig.2023.110747] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
As a hallmark for cancer, aerobic glycolysis, also known as the Warburg effect contributes to tumor progression. However, the roles of aerobic glycolysis on cervical cancer remain elusive. In this work, we identified transcription factor HOXA1 as a novel regulator of aerobic glycolysis. High expression of HOXA1 is closely associated with poor outcome of patients. And, altered HOXA1 expression enhance or reduce aerobic glycolysis and progression in cervical cancer. Mechanistically, HOXA1 directly regulates the transcriptional activity of ENO1 and PGK1, thus induce glycolysis and promote cancer progression. Moreover, therapeutic knockdown of HOXA1 results in reduce aerobic glycolysis and inhibits cervical cancer progression in vivo and in vitro. In conclusion, these data indicate a therapeutic role of HOXA1 inhibits aerobic glycolysis and cervical cancer progression.
Collapse
Affiliation(s)
- Zihui Zhang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jiaxin Peng
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Bingshu Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zhi Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Haoyu Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Ying Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China.
| |
Collapse
|
21
|
Parrillo L, Spinelli R, Longo M, Zatterale F, Santamaria G, Leone A, Campitelli M, Raciti GA, Beguinot F. The Transcription Factor HOXA5: Novel Insights into Metabolic Diseases and Adipose Tissue Dysfunction. Cells 2023; 12:2090. [PMID: 37626900 PMCID: PMC10453582 DOI: 10.3390/cells12162090] [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: 06/15/2023] [Revised: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The transcription factor HOXA5, from the HOX gene family, has long been studied due to its critical role in physiological activities in normal cells, such as organ development and body patterning, and pathological activities in cancer cells. Nonetheless, recent evidence supports the hypothesis of a role for HOXA5 in metabolic diseases, particularly in obesity and type 2 diabetes (T2D). In line with the current opinion that adipocyte and adipose tissue (AT) dysfunction belong to the group of primary defects in obesity, linking this condition to an increased risk of insulin resistance (IR) and T2D, the HOXA5 gene has been shown to regulate adipocyte function and AT remodeling both in humans and mice. Epigenetics adds complexity to HOXA5 gene regulation in metabolic diseases. Indeed, epigenetic mechanisms, specifically DNA methylation, influence the dynamic HOXA5 expression profile. In human AT, the DNA methylation profile at the HOXA5 gene is associated with hypertrophic obesity and an increased risk of developing T2D. Thus, an inappropriate HOXA5 gene expression may be a mechanism causing or maintaining an impaired AT function in obesity and potentially linking obesity to its associated disorders. In this review, we integrate the current evidence about the involvement of HOXA5 in regulating AT function, as well as its association with the pathogenesis of obesity and T2D. We also summarize the current knowledge on the role of DNA methylation in controlling HOXA5 expression. Moreover, considering the susceptibility of epigenetic changes to reversal through targeted interventions, we discuss the potential therapeutic value of targeting HOXA5 DNA methylation changes in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Luca Parrillo
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Rosa Spinelli
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Michele Longo
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Federica Zatterale
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy;
| | - Alessia Leone
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Michele Campitelli
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Gregory Alexander Raciti
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| | - Francesco Beguinot
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (R.S.); (M.L.); (F.Z.); (A.L.); (M.C.); (G.A.R.)
| |
Collapse
|
22
|
Zheng ZQ, Yuan GQ, Zhang GG, Nie QQ, Wang Z. Development and validation of a predictive model in diagnosis and prognosis of primary glioblastoma patients based on Homeobox A family. Discov Oncol 2023; 14:108. [PMID: 37351805 DOI: 10.1007/s12672-023-00726-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Homeobox A (HOXA) family is involved in the development of malignancies as either tumor suppressors or oncogenes. However, their roles in glioblastoma (GBM) and clinical significance have not been fully elucidated. METHODS HOXA mutation and expressions in pan-cancers were investigated using GSCA and Oncomine, which in GBM were validated by cBioPortal, Chinese Glioma Genome Atlas (CGGA), and The Cancer Genome Atlas (TCGA) datasets. Kaplan-Meier analyses were conducted to determine prognostic values of HOXAs at genetic and mRNA levels. Diagnostic roles of HOXAs in tumor classification were explored by GlioVis and R software. Independent prognostic HOXAs were identified using Cox survival analyses, the least absolute shrinkage and selection operator (LASSO) regression, quantitative real-time PCR, and immunohistochemical staining. A HOXAs-based nomogram survival prediction model was developed and evaluated using Kaplan-Meier analysis, time-dependent Area Under Curve, calibration plots, and Decision Curve Analysis in training and validation cohorts. RESULTS HOXAs were highly mutated and overexpressed in pan-cancers, especially in CGGA and TCGA GBM datasets. Genetic alteration and mRNA expression of HOXAs were both found to be prognostic. Specific HOXAs could distinguish IDH mutation (HOXA1-7, HOXA9, HOXA13) and molecular GBM subtypes (HOXA1-2, HOXA9-11, HOXA13). HOXA1/2/3/10 were confirmed to be independent prognostic members, with high expressions validated in clinical GBM tissues. The HOXAs-based nomogram model exhibited good prediction performance and net benefits for patients in training and validation cohorts. CONCLUSION HOXA family has diagnostic values, and the HOXAs-based nomogram model is effective in survival prediction, providing a novel approach to support the treatment of GBM patients.
Collapse
Affiliation(s)
- Zong-Qing Zheng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Gui-Qiang Yuan
- Beijing Neurosurgical Institute & Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Capital Medical University, Beijing, China
| | - Guo-Guo Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Qian-Qian Nie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
23
|
Azuma K, Sakamoto M, Katayama S, Matsui A, Nakamichi K, Goshima N, Watanabe S, Nakayama J, Semba K. HOXB7 induces STAT3-mediated transformation and lung metastasis in immortalized mammary gland NMuMG cells. Genes Cells 2023; 28:277-287. [PMID: 36659836 DOI: 10.1111/gtc.13009] [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: 05/21/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
The homeobox family genes are often dysregulated in various cancer types. Particularly HOXB7 amplification and overexpression correlate with poor prognosis in various cancer such as gastric, pancreatic, and lung cancers. Moreover, HOXB7 is known to contribute to cancer progression by promoting epithelial to mesenchymal transition, anticancer drug resistance, and angiogenesis. In this study, we show that HOXB7 is coamplified with ERBB2 in a subset of breast cancer patients and HOXB7 expression correlates with poor prognosis in HER2-positive breast cancer patients. This clinical observation is supported by the following results-HOXB7 overexpression in an immortalized murine mammary gland epithelial cell line NMuMG induces cellular transformation in vitro, tumorigenesis, and lung metastasis through the activation of JAK-STAT signaling.
Collapse
Affiliation(s)
- Kazushi Azuma
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Mai Sakamoto
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Computational Bio-Big Data Open Innovation Lab (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Shota Katayama
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Atsuka Matsui
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Kazuya Nakamichi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Naoki Goshima
- Division of Transcriptome Analysis, Translational Research Center, Fukushima Medical University, Fukushima, Japan.,Functional Proteomics Team, Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan.,Department of Human Sciences, Musashino University, Tokyo, Japan
| | - Shinya Watanabe
- Translational Research Center, Fukushima Medical University, Fukushima, Japan
| | - Jun Nakayama
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Computational Bio-Big Data Open Innovation Lab (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan.,Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Translational Research Center, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
24
|
Hodkinson LJ, Smith C, Comstra HS, Albanese EH, Ajani BA, Arsalan K, Daisson AP, Forrest KB, Fox EH, Guerette MR, Khan S, Koenig MP, Lam S, Lewandowski AS, Mahoney LJ, Manai N, Miglay J, Miller BA, Milloway O, Ngo VD, Oey NF, Punjani TA, SiMa H, Zeng H, Schmidt CA, Rieder LE. A bioinformatics screen reveals Hox and chromatin remodeling factors at the Drosophila histone locus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.523008. [PMID: 36711759 PMCID: PMC9881919 DOI: 10.1101/2023.01.06.523008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cells orchestrate histone biogenesis with strict temporal and quantitative control. To efficiently regulate histone biogenesis, the repetitive Drosophila melanogaster replication-dependent histone genes are arrayed and clustered at a single locus. Regulatory factors concentrate in a nuclear body known as the histone locus body (HLB), which forms around the locus. Historically, HLB factors are largely discovered by chance, and few are known to interact directly with DNA. It is therefore unclear how the histone genes are specifically targeted for unique and coordinated regulation. To expand the list of known HLB factors, we performed a candidate-based screen by mapping 30 publicly available ChIP datasets and 27 factors to the Drosophila histone gene array. We identified novel transcription factor candidates, including the Drosophila Hox proteins Ultrabithorax, Abdominal-A and Abdominal-B, suggesting a new pathway for these factors in influencing body plan morphogenesis. Additionally, we identified six other transcription factors that target the histone gene array: JIL-1, Hr78, the long isoform of fs(1)h as well as the generalized transcription factors TAF-1, TFIIB, and TFIIF. Our foundational screen provides several candidates for future studies into factors that may influence histone biogenesis. Further, our study emphasizes the powerful reservoir of publicly available datasets, which can be mined as a primary screening technique.
Collapse
Affiliation(s)
- Lauren J Hodkinson
- Genetics and Molecular Biology graduate program, Emory University, Atlanta, GA 30322, USA
| | - Connor Smith
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - H Skye Comstra
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Eric H Albanese
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Bukola A Ajani
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Kawsar Arsalan
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | | | - Katherine B Forrest
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Elijah H Fox
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Matthew R Guerette
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Samia Khan
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Madeleine P Koenig
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Shivani Lam
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Ava S Lewandowski
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Lauren J Mahoney
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Nasserallah Manai
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - JonCarlo Miglay
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Blake A Miller
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Olivia Milloway
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Vu D Ngo
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Nicole F Oey
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Tanya A Punjani
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - HaoMin SiMa
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Hollis Zeng
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Casey A Schmidt
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| | - Leila E Rieder
- Department of Biology 1510 Clifton Road Atlanta, Emory University GA 30322, USA
| |
Collapse
|
25
|
Abstract
Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms. Hox genes are unique because they are spatially and temporally regulated during development in a manner that is dictated by their tightly linked genomic organization. Although their genetic function during embryonic development has been interrogated, less is known about how these transcription factors regulate downstream genes to direct morphogenetic events. Moreover, the continued expression and function of Hox genes at postnatal and adult stages highlights crucial roles for these genes throughout the life of an organism. Here, we provide an overview of Hox genes, highlighting their evolutionary history, their unique genomic organization and how this impacts the regulation of their expression, what is known about their protein structure, and their deployment in development and beyond.
Collapse
Affiliation(s)
- Katharine A. Hubert
- Program in Genetics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
26
|
The pioneering function of the hox transcription factors. Semin Cell Dev Biol 2022:S1084-9521(22)00354-8. [PMID: 36517345 DOI: 10.1016/j.semcdb.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Ever since the discovery that the Hox family of transcription factors establish morphological diversity in the developing embryo, major efforts have been directed towards understanding Hox-dependent patterning. This has led to important discoveries, notably on the mechanisms underlying the collinear expression of Hox genes and Hox binding specificity. More recently, several studies have provided evidence that Hox factors have the capacity to bind their targets in an inaccessible chromatin context and trigger the switch to an accessible, transcriptional permissive, chromatin state. In this review, we provide an overview of the evidences supporting that Hox factors behave as pioneer factors and discuss the potential mechanisms implicated in Hox pioneer activity as well as the significance of this functional property in Hox-dependent patterning.
Collapse
|
27
|
Merabet S, Carnesecchi J. Hox dosage and morphological diversification during development and evolution. Semin Cell Dev Biol 2022:S1084-9521(22)00360-3. [PMID: 36481343 DOI: 10.1016/j.semcdb.2022.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/15/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Hox genes encode for evolutionary conserved transcription factors that have long fascinated biologists since the observation of the first homeotic transformations in flies. Hox genes are developmental architects that instruct the formation of various and precise morphologies along the body axes in cnidarian and bilaterian species. In contrast to these highly specific developmental functions, Hox genes encode for proteins that display poorly selective DNA-binding properties in vitro. This "Hox paradox" has been partially solved with the discovery of the TALE-class cofactors, which interact with all Hox members and form versatile Hox/TALE protein complexes on DNA. Here, we describe the role of the Hox dosage as an additional molecular strategy contributing to further resolve the Hox paradox. We present several cases where the Hox dosage is involved in the formation of different morphologies in invertebrates and vertebrates, with a particular emphasis on flight appendages in insects. We also discuss how the Hox dosage could be interpreted in different types of target enhancers within the nuclear environment in vivo. Altogether our survey underlines the Hox dosage as a key mechanism for shaping Hox molecular function during development and evolution.
Collapse
|
28
|
Bioinformatic Analyses of Broad H3K79me2 Domains in Different Leukemia Cell Line Data Sets. Cells 2022; 11:cells11182830. [PMID: 36139405 PMCID: PMC9496709 DOI: 10.3390/cells11182830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
A subset of expressed genes is associated with a broad H3K4me3 (histone H3 trimethylated at lysine 4) domain that extends throughout the gene body. Genes marked in this way in normal cells are involved in cell-identity and tumor-suppressor activities, whereas in cancer cells, genes driving the cancer phenotype (oncogenes) have this feature. Other histone modifications associated with expressed genes that display a broad domain have been less studied. Here, we identified genes with the broadest H3K79me2 (histone H3 dimethylated at lysine 79) domain in human leukemic cell lines representing different forms of leukemia. Taking a bioinformatic approach, we provide evidence that genes with the broadest H3K79me2 domain have known roles in leukemia (e.g., JMJD1C). In the mixed-lineage leukemia cell line MOLM-13, the HOXA9 gene is in a 100 kb broad H3K79me2 domain with other HOXA protein-coding and oncogenic long non-coding RNA genes. The genes in this domain contribute to leukemia. This broad H3K79me2 domain has an unstable chromatin structure, as was evident by enhanced chromatin accessibility throughout. Together, we provide evidence that identification of genes with the broadest H3K79me2 domain will aid in generating a panel of genes in the diagnosis and therapeutic treatment of leukemia in the future.
Collapse
|
29
|
Yu Z, Liu Z, Lian X, Cheng X, Liu B, Zhang B, Wang H, Wang J, Li A, Ren Z, Pang B, Qian R, Gao Y. High expression of HOXA4 in patients with glioma indicates unfavorable clinical outcomes. Cell Cycle 2022; 21:2387-2402. [PMID: 35852388 DOI: 10.1080/15384101.2022.2096715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Zhenghong Yu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Xiaoyu Lian
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan Zhengzhou, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Binfeng Liu
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan Zhengzhou, China
| | - Bo Zhang
- Department of Surgery of Spine and Spinal Cord, Henan University People’s Hospital, Henan Provincial People’s Hospital, Henan Zhengzhou, China
| | - Hongbo Wang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Jialin Wang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Ang Li
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| | - Zhishuai Ren
- Department of Surgery of Spine and Spinal Cord, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan Zhengzhou, China
| | - Bo Pang
- Department of Neurosurgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, P.R.China
| | - Rongjun Qian
- Department of Neurosurgery, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Henan Province Intelligent Orthopedic Technology Innovation and Transformation International Joint Laboratory, Henan Key Laboratory for Intelligent Precision Orthopedics, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Henan Zhengzhou, China
| |
Collapse
|
30
|
Belpaire M, Taminiau A, Geerts D, Rezsohazy R. HOXA1, a breast cancer oncogene. Biochim Biophys Acta Rev Cancer 2022; 1877:188747. [PMID: 35675857 DOI: 10.1016/j.bbcan.2022.188747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
More than 25 years ago, the first literature records mentioned HOXA1 expression in human breast cancer. A few years later, HOXA1 was confirmed as a proper oncogene in mammary tissue. In the following two decades, molecular data about the mode of action of the HOXA1 protein, the factors contributing to activate and maintain HOXA1 gene expression and the identity of its target genes have accumulated and provide a wider view on the association of this transcription factor to breast oncogenesis. Large-scale transcriptomic data gathered from wide cohorts of patients further allowed refining the relationship between breast cancer type and HOXA1 expression. Several recent reports have reviewed the connection between cancer hallmarks and the biology of HOX genes in general. Here we take HOXA1 as a paradigm and propose an extensive overview of the molecular data centered on this oncoprotein, from what its expression modulators, to the interactors contributing to its oncogenic activities, and to the pathways and genes it controls. The data converge to an intricate picture that answers questions on the multi-modality of its oncogene activities, point towards better understanding of breast cancer aetiology and thereby provides an appraisal for treatment opportunities.
Collapse
Affiliation(s)
- Magali Belpaire
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Dirk Geerts
- Heart Failure Research Center, Amsterdam University Medical Center (AMC), Universiteit van Amsterdam, Amsterdam, the Netherlands.
| | - René Rezsohazy
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium.
| |
Collapse
|
31
|
Perez CJ, Mecklenburg L, Fernandez A, Cantero M, de Souza TA, Lin K, Dent SY, Montoliu L, Awgulewitsch A, Benavides F. Naked (N) mutant mice carry a nonsense mutation in the homeobox of Hoxc13. Exp Dermatol 2022; 31:330-340. [PMID: 34657330 PMCID: PMC11892394 DOI: 10.1111/exd.14469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/23/2021] [Accepted: 10/12/2021] [Indexed: 11/28/2022]
Abstract
Loss of function mutations in HOXC13 have been associated with Ectodermal Dysplasia-9, Hair/Nail Type (ECTD9) in consanguineous families, characterized by sparse to complete absence of hair and nail dystrophy. Here we characterize the spontaneous mouse mutation Naked (N) as a terminal truncation in the Hoxc13 (homeobox C13) gene. Similar to previous reports for homozygous Hoxc13 knock-out (KO) mice, homozygous N/N mice exhibit generalized alopecia with abnormal nails and a short lifespan. However, in contrast to Hoxc13 heterozygous KO mice, N/+ mice show generalized or partial alopecia, associated with loss of hair fibres, along with normal lifespan and fertility. Our data point to a lack of nonsense-mediated Hoxc13 transcript decay and the presence of the truncated mutant protein in N/N and N/+ hair follicles, thus suggesting a dominant-negative mutation. To our knowledge, this is the first report of a semi-dominant and potentially dominant-negative mutation affecting Hoxc13/HOXC13. Furthermore, recreating the N mutant allele in mice using CRISPR/Cas9-mediated genome editing resulted in the same spectrum of deficiencies as those associated with the spontaneous Naked mutation, thus confirming that N is indeed a Hoxc13 mutant allele. Considering the low viability of the Hoxc13 KO mice, the Naked mutation provides an attractive new model for studying ECTD9 disease mechanisms.
Collapse
Affiliation(s)
- Carlos J. Perez
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | | | - Almudena Fernandez
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC) Madrid, Spain and CIBERER-ISCIII, Madrid, Spain
| | - Marta Cantero
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC) Madrid, Spain and CIBERER-ISCIII, Madrid, Spain
| | - Tiago Antonio de Souza
- Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374 - ICB II, São Paulo - SP, Brazil
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Sharon Y.R. Dent
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, Texas, USA
| | - Lluis Montoliu
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC) Madrid, Spain and CIBERER-ISCIII, Madrid, Spain
| | - Alexander Awgulewitsch
- Department of Medicine and Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina (MUSC), 173 Ashley Avenue, CRI 606, Charleston, SC 29425
| | - Fernando Benavides
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, Texas, USA
| |
Collapse
|
32
|
Shenoy US, Adiga D, Kabekkodu SP, Hunter KD, Radhakrishnan R. Molecular implications of HOX genes targeting multiple signaling pathways in cancer. Cell Biol Toxicol 2022; 38:1-30. [PMID: 34617205 PMCID: PMC8789642 DOI: 10.1007/s10565-021-09657-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022]
Abstract
Homeobox (HOX) genes encode highly conserved homeotic transcription factors that play a crucial role in organogenesis and tissue homeostasis. Their deregulation impacts the function of several regulatory molecules contributing to tumor initiation and progression. A functional bridge exists between altered gene expression of individual HOX genes and tumorigenesis. This review focuses on how deregulation in the HOX-associated signaling pathways contributes to the metastatic progression in cancer. We discuss their functional significance, clinical implications and ascertain their role as a diagnostic and prognostic biomarker in the various cancer types. Besides, the mechanism of understanding the theoretical underpinning that affects HOX-mediated therapy resistance in cancers has been outlined. The knowledge gained shall pave the way for newer insights into the treatment of cancer.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Keith D Hunter
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, UK
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
33
|
OUP accepted manuscript. Stem Cells 2022; 40:175-189. [DOI: 10.1093/stmcls/sxab014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/28/2021] [Indexed: 11/14/2022]
|
34
|
Bridoux L, Gofflot F, Rezsohazy R. HOX Protein Activity Regulation by Cellular Localization. J Dev Biol 2021; 9:jdb9040056. [PMID: 34940503 PMCID: PMC8707151 DOI: 10.3390/jdb9040056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
While the functions of HOX genes have been and remain extensively studied in distinct model organisms from flies to mice, the molecular biology of HOX proteins remains poorly documented. In particular, the mechanisms involved in regulating the activity of HOX proteins have been poorly investigated. Nonetheless, based on data available from other well-characterized transcription factors, it can be assumed that HOX protein activity must be finely tuned in a cell-type-specific manner and in response to defined environmental cues. Indeed, records in protein–protein interaction databases or entries in post-translational modification registries clearly support that HOX proteins are the targets of multiple layers of regulation at the protein level. In this context, we review here what has been reported and what can be inferred about how the activities of HOX proteins are regulated by their intracellular distribution.
Collapse
|
35
|
Mallen J, Kalsan M, Zarrineh P, Bridoux L, Ahmad S, Bobola N. Molecular Characterization of HOXA2 and HOXA3 Binding Properties. J Dev Biol 2021; 9:jdb9040055. [PMID: 34940502 PMCID: PMC8707757 DOI: 10.3390/jdb9040055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 01/06/2023] Open
Abstract
The highly conserved HOX homeodomain (HD) transcription factors (TFs) establish the identity of different body parts along the antero–posterior axis of bilaterian animals. Segment diversification and the morphogenesis of different structures is achieved by generating precise patterns of HOX expression along the antero–posterior axis and by the ability of different HOX TFs to instruct unique and specific transcriptional programs. However, HOX binding properties in vitro, characterised by the recognition of similar AT-rich binding sequences, do not account for the ability of different HOX to instruct segment-specific transcriptional programs. To address this problem, we previously compared HOXA2 and HOXA3 binding in vivo. Here, we explore if sequence motif enrichments observed in vivo are explained by binding affinities in vitro. Unexpectedly, we found that the highest enriched motif in HOXA2 peaks was not recognised by HOXA2 in vitro, highlighting the importance of investigating HOX binding in its physiological context. We also report the ability of HOXA2 and HOXA3 to heterodimerise, which may have functional consequences for the HOX patterning function in vivo.
Collapse
Affiliation(s)
- Joshua Mallen
- School of Medical Sciences, University of Manchester, Manchester M13 9PT, UK; (J.M.); (P.Z.)
| | - Manisha Kalsan
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (M.K.); (S.A.)
| | - Peyman Zarrineh
- School of Medical Sciences, University of Manchester, Manchester M13 9PT, UK; (J.M.); (P.Z.)
| | - Laure Bridoux
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 5 (L7.07.10) Place Croix du Sud, 1348 Louvain-la-Neuve, Belgium;
| | - Shandar Ahmad
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (M.K.); (S.A.)
| | - Nicoletta Bobola
- School of Medical Sciences, University of Manchester, Manchester M13 9PT, UK; (J.M.); (P.Z.)
- Correspondence:
| |
Collapse
|
36
|
Induction of HOXA3 by PRRSV inhibits IFN-I response through negatively regulation of HO-1 transcription. J Virol 2021; 96:e0186321. [PMID: 34851144 DOI: 10.1128/jvi.01863-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type I interferons (IFN-I) play a key role in the host defense against virus infection, but porcine reproductive and respiratory syndrome virus (PRRSV) infection does not effectively activate IFN-I response, and the underlying molecular mechanisms are poorly characterized. In this study, a novel transcription factor of the heme oxygenase-1 (HO-1) gene, homeobox A3 (HOXA3), was screened and identified. Here, we found that HOXA3 was significantly increased during PRRSV infection. We demonstrated that HOXA3 promotes PRRSV replication by negatively regulating the HO-1 gene transcription, which is achieved by regulating type I interferons (IFN-I) production. A detailed analysis showed that PRRSV exploits HOXA3 to suppress beta interferon (IFN-β) and IFN-stimulated gene (ISG) expression in host cells. We also provide direct evidence that the activation of IFN-I by HO-1 depends on its interaction with IRF3. Then we further proved that deficiency of HOXA3 promoted the HO-1-IRF3 interaction, and subsequently enhanced IRF3 phosphorylation and nuclear translocation in PRRSV-infected cells. These data suggest that PRRSV uses HOXA3 to negatively regulate the transcription of the HO-1 gene to suppress the IFN-I response for immune evasion. IMPORTANCE Porcine reproductive and respiratory syndrome (PRRS), caused by PRRSV, leads the pork industry worldwide to significant economic losses. HOXA3 is generally considered to be an important molecule in the process of body development and cell differentiation. Here, we found a novel transcription factor of the HO-1 gene, HOXA3, can negatively regulate the transcription of the HO-1 gene and play an important role in the suppression of IFN-I response by PRRSV. PRRSV induces the upregulation of HOXA3, which can negatively regulate HO-1 gene transcription, thereby weakening the interaction between HO-1 and IRF3 for inhibiting the type I IFN response. This study extends the function of HOXA3 to the virus field for the first time and provides new insights into PRRSV immune evasion mechanism.
Collapse
|
37
|
Poliacikova G, Maurel-Zaffran C, Graba Y, Saurin AJ. Hox Proteins in the Regulation of Muscle Development. Front Cell Dev Biol 2021; 9:731996. [PMID: 34733846 PMCID: PMC8558437 DOI: 10.3389/fcell.2021.731996] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Hox genes encode evolutionary conserved transcription factors that specify the anterior-posterior axis in all bilaterians. Being well known for their role in patterning ectoderm-derivatives, such as CNS and spinal cord, Hox protein function is also crucial in mesodermal patterning. While well described in the case of the vertebrate skeleton, much less is known about Hox functions in the development of different muscle types. In contrast to vertebrates however, studies in the fruit fly, Drosophila melanogaster, have provided precious insights into the requirement of Hox at multiple stages of the myogenic process. Here, we provide a comprehensive overview of Hox protein function in Drosophila and vertebrate muscle development, with a focus on the molecular mechanisms underlying target gene regulation in this process. Emphasizing a tight ectoderm/mesoderm cross talk for proper locomotion, we discuss shared principles between CNS and muscle lineage specification and the emerging role of Hox in neuromuscular circuit establishment.
Collapse
Affiliation(s)
| | | | - Yacine Graba
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Andrew J Saurin
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| |
Collapse
|
38
|
Belpaire M, Ewbank B, Taminiau A, Bridoux L, Deneyer N, Marchese D, Lima-Mendez G, Baurain JF, Geerts D, Rezsohazy R. HOXA1 Is an Antagonist of ERα in Breast Cancer. Front Oncol 2021; 11:609521. [PMID: 34490074 PMCID: PMC8417444 DOI: 10.3389/fonc.2021.609521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 07/12/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is a heterogeneous disease and the leading cause of female cancer mortality worldwide. About 70% of breast cancers express ERα. HOX proteins are master regulators of embryo development which have emerged as being important players in oncogenesis. HOXA1 is one of them. Here, we present bioinformatic analyses of genome-wide mRNA expression profiles available in large public datasets of human breast cancer samples. We reveal an extremely strong opposite correlation between HOXA1 versus ER expression and that of 2,486 genes, thereby supporting a functional antagonism between HOXA1 and ERα. We also demonstrate in vitro that HOXA1 can inhibit ERα activity. This inhibition is at least bimodal, requiring an intact HOXA1 DNA-binding homeodomain and involving the DNA-binding independent capacity of HOXA1 to activate NF-κB. We provide evidence that the HOXA1-PBX interaction known to be critical for the transcriptional activity of HOXA1 is not involved in the ERα inhibition. Finally, we reveal that HOXA1 and ERα can physically interact but that this interaction is not essential for the HOXA1-mediated inhibition of ERα. Like other HOX oncoproteins interacting with ERα, HOXA1 could be involved in endocrine therapy resistance.
Collapse
Affiliation(s)
- Magali Belpaire
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Bruno Ewbank
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Arnaud Taminiau
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Laure Bridoux
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Noémie Deneyer
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Damien Marchese
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Gipsi Lima-Mendez
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Jean-François Baurain
- Pôle d'imagerie moléculaire, radiothérapie et oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Woluwe-Saint-Lambert, Belgium.,King Albert II Cancer Institute, Cliniques Universitaires St Luc, Woluwe-Saint-Lambert, Belgium
| | - Dirk Geerts
- Department of Medical Biology, Amsterdam University Medical Centrum (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - René Rezsohazy
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| |
Collapse
|
39
|
A TALE/HOX code unlocks WNT signalling response towards paraxial mesoderm. Nat Commun 2021; 12:5136. [PMID: 34446717 PMCID: PMC8390530 DOI: 10.1038/s41467-021-25370-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/04/2021] [Indexed: 12/02/2022] Open
Abstract
One fundamental yet unresolved question in biology remains how cells interpret the same signalling cues in a context-dependent manner resulting in lineage specification. A key step for decoding signalling cues is the establishment of a permissive chromatin environment at lineage-specific genes triggering transcriptional responses to inductive signals. For instance, bipotent neuromesodermal progenitors (NMPs) are equipped with a WNT-decoding module, which relies on TCFs/LEF activity to sustain both NMP expansion and paraxial mesoderm differentiation. However, how WNT signalling activates lineage specific genes in a temporal manner remains unclear. Here, we demonstrate that paraxial mesoderm induction relies on the TALE/HOX combinatorial activity that simultaneously represses NMP genes and activates the differentiation program. We identify the BRACHYURY-TALE/HOX code that destabilizes the nucleosomes at WNT-responsive regions and establishes the permissive chromatin landscape for de novo recruitment of the WNT-effector LEF1, unlocking the WNT-mediated transcriptional program that drives NMPs towards the paraxial mesodermal fate. Cells in the developing embryo interpret WNT signalling with context-dependence, but the mechanism decoding these cues is unclear. Here, the authors show that combinatorial TALE/HOX activity destabilizes nucleosomes at WNT-responsive regions to activate paraxial mesodermal genes.
Collapse
|
40
|
Hernández R, Jiménez-Luna C, Ortiz R, Setién F, López M, Perazzoli G, Esteller M, Berdasco M, Prados J, Melguizo C. Impact of the Epigenetically Regulated Hoxa-5 Gene in Neural Differentiation from Human Adipose-Derived Stem Cells. BIOLOGY 2021; 10:biology10080802. [PMID: 34440035 PMCID: PMC8389620 DOI: 10.3390/biology10080802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022]
Abstract
Human adipose-derived mesenchymal stem cells (hASCs) may be used in some nervous system pathologies, although obtaining an adequate degree of neuronal differentiation is an important barrier to their applicability. This requires a deep understanding of the expression and epigenetic changes of the most important genes involved in their differentiation. We used hASCs from human lipoaspirates to induce neuronal-like cells through three protocols (Neu1, 2, and 3), determined the degree of neuronal differentiation using specific biomarkers in culture cells and neurospheres, and analyzed epigenetic changes of genes involved in this differentiation. Furthermore, we selected the Hoxa-5 gene to determine its potential to improve neuronal differentiation. Our results showed that an excellent hASC neuronal differentiation process using Neu1 which efficiently modulated NES, CHAT, SNAP25, or SCN9A neuronal marker expression. In addition, epigenetic studies showed relevant changes in Hoxa-5, GRM4, FGFR1, RTEL1, METRN, and PAX9 genes. Functional studies of the Hoxa-5 gene using CRISPR/dCas9 and lentiviral systems showed that its overexpression induced hASCs neuronal differentiation that was accelerated with the exposure to Neu1. These results suggest that Hoxa-5 is an essential gene in hASCs neuronal differentiation and therefore, a potential candidate for the development of cell therapy strategies in neurological disorders.
Collapse
Affiliation(s)
- Rosa Hernández
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Cristina Jiménez-Luna
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Raúl Ortiz
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Fernando Setién
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (F.S.); (M.L.); (M.E.); (M.B.)
- Cancer Epigenetics Group, Cancer and Leukemia Epigenetics and Biology Program (PEBCL), Josep Carreras Leukemia Research Institute (IJC), 08916 Barcelona, Spain
| | - Miguel López
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (F.S.); (M.L.); (M.E.); (M.B.)
- Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Gloria Perazzoli
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (F.S.); (M.L.); (M.E.); (M.B.)
- Cancer Epigenetics Group, Cancer and Leukemia Epigenetics and Biology Program (PEBCL), Josep Carreras Leukemia Research Institute (IJC), 08916 Barcelona, Spain
| | - María Berdasco
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (F.S.); (M.L.); (M.E.); (M.B.)
- Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Jose Prados
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Correspondence:
| | - Consolación Melguizo
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain; (R.H.); (C.J.-L.); (R.O.); (G.P.); (C.M.)
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| |
Collapse
|
41
|
Wilmerding A, Bouteille L, Rinaldi L, Caruso N, Graba Y, Delfini MC. HOXB8 Counteracts MAPK/ERK Oncogenic Signaling in a Chicken Embryo Model of Neoplasia. Int J Mol Sci 2021; 22:8911. [PMID: 34445617 PMCID: PMC8396257 DOI: 10.3390/ijms22168911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
HOX transcription factors are members of an evolutionarily conserved family of proteins required for the establishment of the anteroposterior body axis during bilaterian development. Although they are often deregulated in cancers, the molecular mechanisms by which they act as oncogenes or tumor suppressor genes are only partially understood. Since the MAPK/ERK signaling pathway is deregulated in most cancers, we aimed at apprehending if and how the Hox proteins interact with ERK oncogenicity. Using an in vivo neoplasia model in the chicken embryo consisting in the overactivation of the ERK1/2 kinases in the trunk neural tube, we analyzed the consequences of the HOXB8 gain of function at the morphological and transcriptional levels. We found that HOXB8 acts as a tumor suppressor, counteracting ERK-induced neoplasia. The HOXB8 tumor suppressor function relies on a large reversion of the oncogenic transcriptome induced by ERK. In addition to showing that the HOXB8 protein controls the transcriptional responsiveness to ERK oncogenic signaling, our study identified new downstream targets of ERK oncogenic activation in an in vivo context that could provide clues for therapeutic strategies.
Collapse
Affiliation(s)
- Axelle Wilmerding
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Lauranne Bouteille
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Lucrezia Rinaldi
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
- Beth Israel Deaconess Medical Center, Department of Medicine and the Cancer Center, Division of Hematology, Harvard Initiative of RNA Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Caruso
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Yacine Graba
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Marie-Claire Delfini
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| |
Collapse
|
42
|
Jin L, Tang Q, Hu S, Chen Z, Zhou X, Zeng B, Wang Y, He M, Li Y, Gui L, Shen L, Long K, Ma J, Wang X, Chen Z, Jiang Y, Tang G, Zhu L, Liu F, Zhang B, Huang Z, Li G, Li D, Gladyshev VN, Yin J, Gu Y, Li X, Li M. A pig BodyMap transcriptome reveals diverse tissue physiologies and evolutionary dynamics of transcription. Nat Commun 2021; 12:3715. [PMID: 34140474 PMCID: PMC8211698 DOI: 10.1038/s41467-021-23560-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
A comprehensive transcriptomic survey of pigs can provide a mechanistic understanding of tissue specialization processes underlying economically valuable traits and accelerate their use as a biomedical model. Here we characterize four transcript types (lncRNAs, TUCPs, miRNAs, and circRNAs) and protein-coding genes in 31 adult pig tissues and two cell lines. We uncover the transcriptomic variability among 47 skeletal muscles, and six adipose depots linked to their different origins, metabolism, cell composition, physical activity, and mitochondrial pathways. We perform comparative analysis of the transcriptomes of seven tissues from pigs and nine other vertebrates to reveal that evolutionary divergence in transcription potentially contributes to lineage-specific biology. Long-range promoter–enhancer interaction analysis in subcutaneous adipose tissues across species suggests evolutionarily stable transcription patterns likely attributable to redundant enhancers buffering gene expression patterns against perturbations, thereby conferring robustness during speciation. This study can facilitate adoption of the pig as a biomedical model for human biology and disease and uncovers the molecular bases of valuable traits. A comprehensive transcriptomic survey of the pig could enable mechanistic understanding of tissue specialization and accelerate its use as a biomedical model. Here the authors characterize four distinct transcript types in 31 adult pig tissues to dissect their distinct structural and transcriptional features and uncover transcriptomic variability related to tissue physiology.
Collapse
Affiliation(s)
- Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China.
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongxu Chen
- Department of Life Science, Tcuni Inc., Chengdu, Sichuan, China
| | - Xuming Zhou
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bo Zeng
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuhao Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mengnan He
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lixuan Gui
- Department of Life Science, Tcuni Inc., Chengdu, Sichuan, China
| | - Linyuan Shen
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Keren Long
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jideng Ma
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xun Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhengli Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanzhi Jiang
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Guoqing Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Li Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Fei Liu
- Information and Educational Technology Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Zhang
- Ya'an Digital Economy Operation Company, Ya'an, Sichuan, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guisen Li
- Renal Department and Nephrology Institute, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yiren Gu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Xuewei Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
43
|
Yoshioka K, Nagahisa H, Miura F, Araki H, Kamei Y, Kitajima Y, Seko D, Nogami J, Tsuchiya Y, Okazaki N, Yonekura A, Ohba S, Sumita Y, Chiba K, Ito K, Asahina I, Ogawa Y, Ito T, Ohkawa Y, Ono Y. Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle. SCIENCE ADVANCES 2021; 7:7/24/eabd7924. [PMID: 34108202 PMCID: PMC8189581 DOI: 10.1126/sciadv.abd7924] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 04/21/2021] [Indexed: 05/04/2023]
Abstract
Muscle stem cells (satellite cells) are distributed throughout the body and have heterogeneous properties among muscles. However, functional topographical genes in satellite cells of adult muscle remain unidentified. Here, we show that expression of Homeobox-A (Hox-A) cluster genes accompanied with DNA hypermethylation of the Hox-A locus was robustly maintained in both somite-derived muscles and their associated satellite cells in adult mice, which recapitulates their embryonic origin. Somite-derived satellite cells were clearly separated from cells derived from cranial mesoderm in Hoxa10 expression. Hoxa10 inactivation led to genomic instability and mitotic catastrophe in somite-derived satellite cells in mice and human. Satellite cell-specific Hoxa10 ablation in mice resulted in a decline in the regenerative ability of somite-derived muscles, which were unobserved in cranial mesoderm-derived muscles. Thus, our results show that Hox gene expression profiles instill the embryonic history in satellite cells as positional memory, potentially modulating region-specific pathophysiology in adult muscles.
Collapse
Affiliation(s)
- Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hiroshi Nagahisa
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Daiki Seko
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Jumpei Nogami
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshifumi Tsuchiya
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Narihiro Okazaki
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Akihiko Yonekura
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ko Chiba
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
44
|
Wilmerding A, Rinaldi L, Caruso N, Lo Re L, Bonzom E, Saurin AJ, Graba Y, Delfini MC. HoxB genes regulate neuronal delamination in the trunk neural tube by controlling the expression of Lzts1. Development 2021; 148:dev.195404. [PMID: 33472847 DOI: 10.1242/dev.195404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/11/2021] [Indexed: 01/23/2023]
Abstract
Differential Hox gene expression is central for specification of axial neuronal diversity in the spinal cord. Here, we uncover an additional function of Hox proteins in the developing spinal cord, restricted to B cluster Hox genes. We found that members of the HoxB cluster are expressed in the trunk neural tube of chicken embryo earlier than Hox from the other clusters, with poor antero-posterior axial specificity and with overlapping expression in the intermediate zone (IZ). Gain-of-function experiments of HoxB4, HoxB8 and HoxB9, respectively, representative of anterior, central and posterior HoxB genes, resulted in ectopic progenitor cells in the mantle zone. The search for HoxB8 downstream targets in the early neural tube identified the leucine zipper tumor suppressor 1 gene (Lzts1), the expression of which is also activated by HoxB4 and HoxB9. Gain- and loss-of-function experiments showed that Lzts1, which is expressed endogenously in the IZ, controls neuronal delamination. These data collectively indicate that HoxB genes have a generic function in the developing spinal cord, controlling the expression of Lzts1 and neuronal delamination.
Collapse
Affiliation(s)
| | | | - Nathalie Caruso
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Laure Lo Re
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Emilie Bonzom
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Andrew J Saurin
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Yacine Graba
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | | |
Collapse
|
45
|
Bridoux L, Zarrineh P, Mallen J, Phuycharoen M, Latorre V, Ladam F, Losa M, Baker SM, Sagerstrom C, Mace KA, Rattray M, Bobola N. HOX paralogs selectively convert binding of ubiquitous transcription factors into tissue-specific patterns of enhancer activation. PLoS Genet 2020; 16:e1009162. [PMID: 33315856 PMCID: PMC7769617 DOI: 10.1371/journal.pgen.1009162] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 12/28/2020] [Accepted: 09/28/2020] [Indexed: 11/18/2022] Open
Abstract
Gene expression programs determine cell fate in embryonic development and their dysregulation results in disease. Transcription factors (TFs) control gene expression by binding to enhancers, but how TFs select and activate their target enhancers is still unclear. HOX TFs share conserved homeodomains with highly similar sequence recognition properties, yet they impart the identity of different animal body parts. To understand how HOX TFs control their specific transcriptional programs in vivo, we compared HOXA2 and HOXA3 binding profiles in the mouse embryo. HOXA2 and HOXA3 directly cooperate with TALE TFs and selectively target different subsets of a broad TALE chromatin platform. Binding of HOX and tissue-specific TFs convert low affinity TALE binding into high confidence, tissue-specific binding events, which bear the mark of active enhancers. We propose that HOX paralogs, alone and in combination with tissue-specific TFs, generate tissue-specific transcriptional outputs by modulating the activity of TALE TFs at selected enhancers.
Collapse
Affiliation(s)
- Laure Bridoux
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Peyman Zarrineh
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Joshua Mallen
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Mike Phuycharoen
- Department of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Victor Latorre
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Frank Ladam
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusets, United States of America
| | - Marta Losa
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Syed Murtuza Baker
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
- School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Charles Sagerstrom
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusets, United States of America
| | - Kimberly A. Mace
- School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Magnus Rattray
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Nicoletta Bobola
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Zha J, Lai Q, Deng M, Shi P, Zhao H, Chen Q, Wu H, Xu B. Disruption of CTCF Boundary at HOXA Locus Promote BET Inhibitors' Therapeutic Sensitivity in Acute Myeloid Leukemia. Stem Cell Rev Rep 2020; 16:1280-1291. [PMID: 33057942 DOI: 10.1007/s12015-020-10057-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
Both HOX gene expression and CTCF regulation have been well demonstrated to play a critical role in regulating maintenance of leukemic stem cells (LSCs) that are known to be resistant to BET inhibitor (BETi). To investigate the regulatory role of CTCF boundary in aberrant HOX gene expression and the therapeutic sensitivity of BETi in AML, we employed CRISPR-Cas9 genome editing technology to delete 47 base pairs of the CTCF binding motif which is located between HOXA7 and HOXA9 genes (CBS7/9) in different subtypes of AML with either MLL-rearrangement or NPM1 mutation. Our results revealed that HOXA9 is significantly downregulated in response to the CBS7/9 deletion. Moreover, CBS7/9 boundary deletion sensitized the BETi treatment reaction in both MOLM-13 and OCI-AML3 cells. To further examine whether BETi therapeutic sensitivity in AML is depended on the expression level of the HOXA9 gene, we overexpressed the HOXA9 in the CBS7/9 deleted AML cell lines, which can rescue and restore the resistance to BETi treatment of the CBS7/9 KO cells by activating MAPK signaling pathway. Deletion of CBS7/9 specifically decreased the recruitment of BRD4 and RNA pol II to the posterior HOXA genes, in which, a transcription elongation factor ELL3 is the key factor in regulating HOXA gene transcription monitored by CBS7/9 chromatin boundary. Thus, disruption of CBS7/9 boundary perturbs HOXA9 transcription and regulates BETi sensitivity in AML treatment. Moreover, alteration of CTCF boundaries in the oncogene loci may provide a novel strategy to overcome the drug resistance of LSCs. Graphical abstract.
Collapse
Affiliation(s)
- Jie Zha
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China
| | - Qian Lai
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China
| | - Manman Deng
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China
| | - Pengcheng Shi
- Department of Hematology, Nanfang Hospital. Southern Medical University, Guangzhou, 510515, China
| | - Haijun Zhao
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China
| | - Qinwei Chen
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China
| | - Hua Wu
- Department of Nuclear Medicine, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Bing Xu
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen, 361003, Fujian, China.
- Key Laboratory for Diagnosis and Treatment of Hematological Malignancy of Xiamen, Xiamen, 361003, China.
| |
Collapse
|
47
|
Pai P, Sukumar S. HOX genes and the NF-κB pathway: A convergence of developmental biology, inflammation and cancer biology. Biochim Biophys Acta Rev Cancer 2020; 1874:188450. [PMID: 33049277 DOI: 10.1016/j.bbcan.2020.188450] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/11/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
The roles of HOX transcription factors as oncogenes and tumor suppressor genes, and the NF-KB pathway in chronic inflammation, both leading to cancer are well-established. HOX transcription factors are members of an evolutionarily conserved family of proteins required for anteroposterior body axis patterning during embryonic development, and are often dysregulated in cancer. The NF-KB pathway aids inflammation and immunity but it is also important during embryonic development. It is frequently activated in both solid and hematological malignancies. NF-KB and HOX proteins can influence each other through mutual transcriptional regulation, protein-protein interactions, and regulation of upstream and downstream interactors. These interactions have important implications both in homeostasis and in disease. In this review, we summarize the role of HOX proteins in regulating inflammation in homeostasis and disease- with a particular emphasis on cancer. We also describe the relationship between HOX genes and the NF-KB pathway, and discuss potential therapeutic strategies.
Collapse
Affiliation(s)
- Priya Pai
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America.
| |
Collapse
|
48
|
Gonçalves CS, Le Boiteux E, Arnaud P, Costa BM. HOX gene cluster (de)regulation in brain: from neurodevelopment to malignant glial tumours. Cell Mol Life Sci 2020; 77:3797-3821. [PMID: 32239260 PMCID: PMC11105007 DOI: 10.1007/s00018-020-03508-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/10/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022]
Abstract
HOX genes encode a family of evolutionarily conserved homeodomain transcription factors that are crucial both during development and adult life. In humans, 39 HOX genes are arranged in four clusters (HOXA, B, C, and D) in chromosomes 7, 17, 12, and 2, respectively. During embryonic development, particular epigenetic states accompany their expression along the anterior-posterior body axis. This tightly regulated temporal-spatial expression pattern reflects their relative chromosomal localization, and is critical for normal embryonic brain development when HOX genes are mainly expressed in the hindbrain and mostly absent in the forebrain region. Epigenetic marks, mostly polycomb-associated, are dynamically regulated at HOX loci and regulatory regions to ensure the finely tuned HOX activation and repression, highlighting a crucial epigenetic plasticity necessary for homeostatic development. HOX genes are essentially absent in healthy adult brain, whereas they are detected in malignant brain tumours, namely gliomas, where HOX genes display critical roles by regulating several hallmarks of cancer. Here, we review the major mechanisms involved in HOX genes (de)regulation in the brain, from embryonic to adult stages, in physiological and oncologic conditions. We focus particularly on the emerging causes of HOX gene deregulation in glioma, as well as on their functional and clinical implications.
Collapse
Affiliation(s)
- Céline S Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Elisa Le Boiteux
- Université Clermont Auvergne, CNRS, INSERM-iGReD, Clermont-Ferrand, France
| | - Philippe Arnaud
- Université Clermont Auvergne, CNRS, INSERM-iGReD, Clermont-Ferrand, France
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
49
|
HOXC4 up-regulates NF-κB signaling and promotes the cell proliferation to drive development of human hematopoiesis, especially CD43+ cells. BLOOD SCIENCE 2020; 2:117-128. [PMID: 35400027 PMCID: PMC8974941 DOI: 10.1097/bs9.0000000000000054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/19/2020] [Indexed: 01/05/2023] Open
Abstract
The hematopoietic function of HOXC4 has not been extensively investigated. Our research indicated that induction of HOXC4 in co-culture system from D10 significantly promoted productions of most hematopoietic progenitor cells. CD34−CD43+ cells could be clearly classified into CD34−CD43low and CD34−CD43high sub-populations at D14. The former cells had greater myelogenic potential, and their production was not significantly influenced by induction of HOXC4. By contrast, the latter cells had greater potential to differentiate into megakaryocytes and erythroid cells, and thus had properties of erythroid–megakaryocyte common progenitors, which abundance was increased by ∼2-fold when HOXC4 was induced from D10. For CD34−CD43low, CD34+CD43+, and CD34−CD43high sub-populations, CD43 level served as a natural index for the tendency to undergo hematopoiesis. Induction of HOXC4 from D10 caused more CD43+ cells sustain in S-phase with up-regulation of NF-κB signaling, which could be counteracted by inhibition of NF-κB signaling. These observations suggested that promotion of hematopoiesis by HOXC4 is closely related to NF-κB signaling and a change in cell-cycle status, which containing potential of clinical applications.
Collapse
|
50
|
Bondos SE, Geraldo Mendes G, Jons A. Context-dependent HOX transcription factor function in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 174:225-262. [PMID: 32828467 DOI: 10.1016/bs.pmbts.2020.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During animal development, HOX transcription factors determine the fate of developing tissues to generate diverse organs and appendages. The power of these proteins is striking: mis-expressing a HOX protein causes homeotic transformation of one body part into another. During development, HOX proteins interpret their cellular context through protein interactions, alternative splicing, and post-translational modifications to regulate cell proliferation, cell death, cell migration, cell differentiation, and angiogenesis. Although mutation and/or mis-expression of HOX proteins during development can be lethal, changes in HOX proteins that do not pattern vital organs can result in survivable malformations. In adults, mutation and/or mis-expression of HOX proteins disrupts their gene regulatory networks, deregulating cell behaviors and leading to arthritis and cancer. On the molecular level, HOX proteins are composed of DNA binding homeodomain, and large regions of unstructured, or intrinsically disordered, protein sequence. The primary roles of HOX proteins in arthritis and cancer suggest that mutations associated with these diseases in both the structured and disordered regions of HOX proteins can have substantial functional effects. These insights lead to new questions critical for understanding and manipulating HOX function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Sarah E Bondos
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States.
| | - Gabriela Geraldo Mendes
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Amanda Jons
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| |
Collapse
|