1
|
Wu XM, Zheng SY, Chang MX. Zebrafish as a Model for Investigating Antiviral Innate Immunity. Methods Mol Biol 2025; 2854:221-236. [PMID: 39192133 DOI: 10.1007/978-1-0716-4108-8_22] [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] [Indexed: 08/29/2024]
Abstract
Zebrafish is a widely used model organism in genetics, developmental biology, pathology, and immunology research. Due to their fast reproduction, large numbers, transparent early embryos, and high genetic conservation with the human genome, zebrafish have been used as a model for studying human and fish viral diseases. In particular, the ability to easily perform forward and reverse genetics and lacking a functional adaptive immune response during the early period of development establish the zebrafish as a favored option to assess the functional implication of specific genes in the antiviral innate immune response and the pathogenesis of viral diseases. In this chapter, we detail protocols for the antiviral innate immunity analysis using the zebrafish model, including the generation of gene-overexpression zebrafish, generation of gene-knockout zebrafish by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology, methods of viral infection in zebrafish larvae, analyzing the expression of antiviral genes in zebrafish larvae using qRT-PCR, Western blotting and transcriptome sequencing, and in vivo antiviral assays. These experimental protocols provide effective references for studying the antiviral immune response in the zebrafish model.
Collapse
Affiliation(s)
- Xiao Man Wu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Si Yao Zheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ming Xian Chang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
2
|
Park JW, Cortes LR, Sandoval NP, Baron AG, Vree AR, Fideles HJ, Hansen MR, Lopez JI, Dilday EA, Rashid S, Kammel LG, van Veen JE, Correa SM. Sex-specific thermoregulatory effects of estrogen signaling in Reprimo lineage cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626488. [PMID: 39677630 PMCID: PMC11642856 DOI: 10.1101/2024.12.02.626488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Menopause affects over a million individuals annually and is characterized by variable and declining ovarian hormones. Decreasing estrogen levels impact energy homeostasis and increases the risk of metabolic disorders. Energy expenditure is largely directed towards thermoregulation, which is modulated in part by estrogen receptor (ER) α expressing neurons in the hypothalamus. Whether specific sub-populations of ERα+ neurons control the effects of estrogens on thermogenesis remains poorly understood. This study investigates the function of ERα in neurons that express Rprm (Reprimo), a gene we previously linked to thermoregulation in females. Here, we use a novel ReprimoCre mouse to selectively knock out ERα in Rprm lineage neurons (Reprimo-specific estrogen receptor α KO; RERKO) and report changes in core temperature in female mice, with no changes in body weight, body composition, or food intake. RERKO females have elevated brown adipose tissue (BAT) temperature and lower tail temperature relative to controls, suggesting increased heat production and impaired heat dissipation, respectively. Developmental expression of Rprm was detected in the brain, but not in BAT or white adipose tissue suggesting temperature changes may be mediated by the nervous system. Thus, we next ablated Rprm expressing neurons in the ventrolateral area of the ventromedial nucleus of the hypothalamus (VMHvl) and observed a reduction in core temperature and increased fat mass in ablated female mice relative to controls. Taken together, these results show that estrogen signaling in Rprm expressing cells and VMHvl Rprm neurons are critical for thermoregulation, mainly through the modulation of brown adipose tissue thermogenesis in female, but not male mice.
Collapse
Affiliation(s)
- Jae W. Park
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Laura R. Cortes
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Norma P. Sandoval
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Alejandra G. Baron
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Adriana R. Vree
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Higor J. Fideles
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Mia R. Hansen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Julissa I. Lopez
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Cypress College, Cypress, CA, USA
| | - Elizabeth A. Dilday
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Sakina Rashid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Laura G. Kammel
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - J. Edward van Veen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Stephanie M. Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
3
|
Ye Z, Wang J, Shi W, Zhou Z, Zhang Y, Wang J, Yang H. Reprimo (RPRM) as a Potential Preventive and Therapeutic Target for Radiation-Induced Brain Injury via Multiple Mechanisms. Int J Mol Sci 2023; 24:17055. [PMID: 38069378 PMCID: PMC10707327 DOI: 10.3390/ijms242317055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China; (Z.Y.); (J.W.); (W.S.); (Z.Z.); (Y.Z.); (J.W.)
| |
Collapse
|
4
|
Li Z, Zhou Z, Tian S, Zhang K, An G, Zhang Y, Ma R, Sheng B, Wang T, Yang H, Yang L. RPRM deletion preserves hematopoietic regeneration by promoting EGFR-dependent DNA repair and hematopoietic stem cell proliferation post ionizing radiation. Cell Biol Int 2022; 46:2158-2172. [PMID: 36041213 PMCID: PMC9804513 DOI: 10.1002/cbin.11900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/28/2022] [Accepted: 08/22/2022] [Indexed: 01/05/2023]
Abstract
Reprimo (RPRM), a target gene of p53, is a known tumor suppressor. DNA damage induces RPRM, which triggers p53-dependent G2 arrest by inhibiting cyclin B1/Cdc2 complex activation and promotes DNA damage-induced apoptosis. RPRM negatively regulates ataxia-telangiectasia mutated by promoting its nuclear-cytoplasmic translocation and degradation, thus inhibiting DNA damage. Therefore, RPRM plays a crucial role in DNA damage response. Moreover, the loss of RPRM confers radioresistance in mice, which enables longer survival and less severe intestinal injury after radiation exposure. However, the role of RPRM in radiation-induced hematopoietic system injury remains unknown. Herein, utilizing a RPRM-knockout mouse model, we found that RPRM deletion did not affect steady-state hematopoiesis in mice. However, RPRM knockout significantly alleviated radiation-induced hematopoietic system injury and preserved mouse hematopoietic regeneration in hematopoietic stem cells (HSCs) against radiation-induced DNA damage. Further mechanistic studies showed that RPRM loss significantly increased EGFR expression and phosphorylation in HSCs to activate STAT3 and DNA-PKcs, thus promoting HSC DNA repair and proliferation. These findings reveal the critical role of RPRM in radiation-induced hematopoietic system injury, confirming our hypothesis that RPRM may serve as a novel target for radiation protection.
Collapse
Affiliation(s)
- Zixuan Li
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina,Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Zhou Zhou
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina
| | - Shuaiyu Tian
- Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Kailu Zhang
- Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Gangli An
- Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Yarui Zhang
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina
| | - Renyuxue Ma
- Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Binjie Sheng
- Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Tian Wang
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina,Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| | - Hongying Yang
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina
| | - Lin Yang
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouJiangsuChina,School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouJiangsuChina,Cyrus Tang Medical Institute, Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuChina
| |
Collapse
|
5
|
Muhammad JS, Siddiqui R, Khan NA. COVID-19 and alcohol use disorder: putative differential gene expression patterns that might be associated with neurological complications. Hosp Pract (1995) 2022; 50:189-195. [PMID: 35686663 DOI: 10.1080/21548331.2022.2088183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Several lines of evidence suggest that SARS-CoV-2 invasion of the central nervous system leads to meningitis and encephalopathy syndromes. Additionally, chronic alcoholics were found to be at a higher risk of developing mental health problems and serious neurological manifestations, if exposed to SARS-CoV-2 infection. METHODS Herein, we studied RNA seq data from alcoholics' brain tissue and COVID-19 patient's brain tissue to identify the common differentially expressed genes. RESULTS Overlap analysis depicted the expression of seven genes (GHRL, SLN, VGF, IL1RL1, NPTX2, PDYN, and RPRML) that were significantly upregulated in both groups. Along with these, protein-protein interaction analysis revealed 10 other key molecules with strong interactions with the aforementioned genes. CONCLUSIONS Taken together with the functional effect of these genes, we suggest a strong molecular link between COVID-19-induced severities and neurological impairment in patients suffering from alcohol abuse disorder. These findings emphasize the importance of identifying chronic alcoholism as a risk factor for developing cognitive and memory impairment in COVID-19 patients.
Collapse
Affiliation(s)
- Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Ruqaiyyah Siddiqui
- College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - Naveed Ahmed Khan
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE
| |
Collapse
|
6
|
He Y, Chen Y, Yao L, Wang J, Sha X, Wang Y. The Inflamm-Aging Model Identifies Key Risk Factors in Atherosclerosis. Front Genet 2022; 13:865827. [PMID: 35706446 PMCID: PMC9191626 DOI: 10.3389/fgene.2022.865827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Atherosclerosis, one of the main threats to human life and health, is driven by abnormal inflammation (i.e., chronic inflammation or oxidative stress) during accelerated aging. Many studies have shown that inflamm-aging exerts a significant impact on the occurrence of atherosclerosis, particularly by inducing an immune homeostasis imbalance. However, the potential mechanism by which inflamm-aging induces atherosclerosis needs to be studied more thoroughly, and there is currently a lack of powerful prediction models.Methods: First, an improved inflamm-aging prediction model was constructed by integrating aging, inflammation, and disease markers with the help of machine learning methods; then, inflamm-aging scores were calculated. In addition, the causal relationship between aging and disease was identified using Mendelian randomization. A series of risk factors were also identified by causal analysis, sensitivity analysis, and network analysis.Results: Our results revealed an accelerated inflamm-aging pattern in atherosclerosis and suggested a causal relationship between inflamm-aging and atherosclerosis. Mechanisms involving inflammation, nutritional balance, vascular homeostasis, and oxidative stress were found to be driving factors of atherosclerosis in the context of inflamm-aging.Conclusion: In summary, we developed a model integrating crucial risk factors in inflamm-aging and atherosclerosis. Our computation pipeline could be used to explore potential mechanisms of related diseases.
Collapse
Affiliation(s)
- Yudan He
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Yao Chen
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Lilin Yao
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Junyi Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Yin Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yin Wang,
| |
Collapse
|
7
|
Alarcón MA, Olivares W, Córdova-Delgado M, Muñoz-Medel M, de Mayo T, Carrasco-Aviño G, Wichmann I, Landeros N, Amigo J, Norero E, Villarroel-Espíndola F, Riquelme A, Garrido M, Owen GI, Corvalán AH. The Reprimo-Like Gene Is an Epigenetic-Mediated Tumor Suppressor and a Candidate Biomarker for the Non-Invasive Detection of Gastric Cancer. Int J Mol Sci 2020; 21:ijms21249472. [PMID: 33322837 PMCID: PMC7763358 DOI: 10.3390/ijms21249472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022] Open
Abstract
Reprimo-like (RPRML) is an uncharacterized member of the Reprimo gene family. Here, we evaluated the role of RPRML and whether its regulation by DNA methylation is a potential non-invasive biomarker of gastric cancer. RPRML expression was evaluated by immunohistochemistry in 90 patients with gastric cancer and associated with clinicopathologic characteristics and outcomes. The role of RPRML in cancer biology was investigated in vitro, through RPRML ectopic overexpression. Functional experiments included colony formation, soft agar, MTS, and Ki67 immunofluorescence assays. DNA methylation-mediated silencing was evaluated by the 5-azacytidine assay and direct bisulfite sequencing. Non-invasive detection of circulating methylated RPRML DNA was assessed in 25 gastric cancer cases and 25 age- and sex-balanced cancer-free controls by the MethyLight assay. Downregulation of RPRML protein expression was associated with poor overall survival in advanced gastric cancer. RPRML overexpression significantly inhibited clonogenic capacity, anchorage-independent growth, and proliferation in vitro. Circulating methylated RPRML DNA distinguished patients with gastric cancer from controls with an area under the curve of 0.726. The in vitro overexpression results and the poor patient survival associated with lower RPRML levels suggest that RPRML plays a tumor-suppressive role in the stomach. Circulating methylated RPRML DNA may serve as a biomarker for the non-invasive detection of gastric cancer.
Collapse
Affiliation(s)
- María Alejandra Alarcón
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
| | - Wilda Olivares
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
| | - Miguel Córdova-Delgado
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
| | - Matías Muñoz-Medel
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
| | - Tomas de Mayo
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
- Faculty of Sciences, School of Medicine Universidad Mayor, Santiago 8580745, Chile
| | - Gonzalo Carrasco-Aviño
- Department of Pathology, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile;
- Department of Pathology, Clínica Las Condes, Santiago 7591210, Chile
| | - Ignacio Wichmann
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
- Department of Obstetrics, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Natalia Landeros
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
| | - Julio Amigo
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile;
| | - Enrique Norero
- Esophagogastric Surgery Unit, Hospital Dr Sótero del Río, Santiago 8207257, Chile;
- Digestive Surgery Department, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Franz Villarroel-Espíndola
- Translational Medicine Laboratory, Instituto Oncológico Fundación Arturo López Pérez (FALP), Santiago 8320000, Chile;
| | - Arnoldo Riquelme
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
| | - Marcelo Garrido
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago 8330005, Chile;
| | - Alejandro H. Corvalán
- Department of Hematology & Oncology, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (M.A.A.); (W.O.); (M.C.-D.); (M.M.-M.); (I.W.); (N.L.); (M.G.)
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8330034, Chile; (T.d.M.); (G.I.O.)
- Correspondence:
| |
Collapse
|
8
|
Gupta MK, Rajeswari J, Reddy PR, Kumar KS, Chamundeswaramma KV, Vadde R. Genetic Marker Identification for the Detection of Early-Onset Gastric Cancer Through Genome-Wide Association Studies. RECENT ADVANCEMENTS IN BIOMARKERS AND EARLY DETECTION OF GASTROINTESTINAL CANCERS 2020:191-211. [DOI: https:/doi.org/10.1007/978-981-15-4431-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
9
|
Gupta MK, Rajeswari J, Reddy PR, Kumar KS, Chamundeswaramma KV, Vadde R. Genetic Marker Identification for the Detection of Early-Onset Gastric Cancer Through Genome-Wide Association Studies. RECENT ADVANCEMENTS IN BIOMARKERS AND EARLY DETECTION OF GASTROINTESTINAL CANCERS 2020:191-211. [DOI: 10.1007/978-981-15-4431-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
10
|
Stanic K, Reig G, Figueroa RJ, Retamal PA, Wichmann IA, Opazo JC, Owen GI, Corvalán AH, Concha ML, Amigo JD. The Reprimo gene family member, reprimo-like (rprml), is required for blood development in embryonic zebrafish. Sci Rep 2019; 9:7131. [PMID: 31073223 PMCID: PMC6509255 DOI: 10.1038/s41598-019-43436-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/24/2019] [Indexed: 11/09/2022] Open
Abstract
The Reprimo gene family comprises a group of single-exon genes for which their physiological function remains poorly understood. Heretofore, mammalian Reprimo (RPRM) has been described as a putative p53-dependent tumor suppressor gene that functions at the G2/M cell cycle checkpoint. Another family member, Reprimo-like (RPRML), has not yet an established role in physiology or pathology. Importantly, RPRML expression pattern is conserved between zebrafish and human species. Here, using CRISPR-Cas9 and antisense morpholino oligonucleotides, we disrupt the expression of rprml in zebrafish and demonstrate that its loss leads to impaired definitive hematopoiesis. The formation of hemangioblasts and the primitive wave of hematopoiesis occur normally in absence of rprml. Later in development there is a significant reduction in erythroid-myeloid precursors (EMP) at the posterior blood island (PBI) and a significant decline of definitive hematopoietic stem/progenitor cells (HSPCs). Furthermore, loss of rprml also increases the activity of caspase-3 in endothelial cells within the caudal hematopoietic tissue (CHT), the first perivascular niche where HSPCs reside during zebrafish embryonic development. Herein, we report an essential role for rprml during hematovascular development in zebrafish embryos, specifically during the definitive waves of hematopoiesis, indicating for the first time a physiological role for the rprml gene.
Collapse
Affiliation(s)
- Karen Stanic
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - German Reig
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Universidad Bernardo O´Higgins, Escuela de Tecnología Médica and Centro Integrativo de Biología y Química Aplicada (CIBQA), Santiago, Chile
| | - Ricardo J Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro A Retamal
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio A Wichmann
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Laboratorio de Oncología, Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Gareth I Owen
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Laboratorio de Oncología, Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel L Concha
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile, Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Julio D Amigo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
11
|
Amigo JD, Opazo JC, Jorquera R, Wichmann IA, Garcia-Bloj BA, Alarcon MA, Owen GI, Corvalán AH. The Reprimo Gene Family: A Novel Gene Lineage in Gastric Cancer with Tumor Suppressive Properties. Int J Mol Sci 2018; 19:E1862. [PMID: 29941787 PMCID: PMC6073456 DOI: 10.3390/ijms19071862] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 12/18/2022] Open
Abstract
The reprimo (RPRM) gene family is a group of single exon genes present exclusively within the vertebrate lineage. Two out of three members of this family are present in humans: RPRM and RPRM-Like (RPRML). RPRM induces cell cycle arrest at G2/M in response to p53 expression. Loss-of-expression of RPRM is related to increased cell proliferation and growth in gastric cancer. This evidence suggests that RPRM has tumor suppressive properties. However, the molecular mechanisms and signaling partners by which RPRM exerts its functions remain unknown. Moreover, scarce studies have attempted to characterize RPRML, and its functionality is unclear. Herein, we highlight the role of the RPRM gene family in gastric carcinogenesis, as well as its potential applications in clinical settings. In addition, we summarize the current knowledge on the phylogeny and expression patterns of this family of genes in embryonic zebrafish and adult humans. Strikingly, in both species, RPRM is expressed primarily in the digestive tract, blood vessels and central nervous system, supporting the use of zebrafish for further functional characterization of RPRM. Finally, drawing on embryonic and adult expression patterns, we address the potential relevance of RPRM and RPRML in cancer. Active investigation or analytical research in the coming years should contribute to novel translational applications of this poorly understood gene family as potential biomarkers and development of novel cancer therapies.
Collapse
Affiliation(s)
- Julio D Amigo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025 Santiago, Chile.
| | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, 5090000 Valdivia, Chile.
| | - Roddy Jorquera
- CORE Biodata, Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile.
| | - Ignacio A Wichmann
- Laboratory of Oncology, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- Departamento de Oncología y Hematología, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- CORE Biodata, Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile.
| | - Benjamin A Garcia-Bloj
- Laboratory of Oncology, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
| | - Maria Alejandra Alarcon
- Laboratory of Oncology, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- Departamento de Oncología y Hematología, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
| | - Gareth I Owen
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025 Santiago, Chile.
- Laboratory of Oncology, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| | - Alejandro H Corvalán
- Laboratory of Oncology, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- Departamento de Oncología y Hematología, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330034 Santiago, Chile.
- CORE Biodata, Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile.
| |
Collapse
|
12
|
Stanic K, Quiroz A, Lemus CG, Wichmann IA, Corvalán AH, Owen GI, Opazo JC, Concha ML, Amigo JD. Expression of RPRM/rprm in the Olfactory System of Embryonic Zebrafish ( Danio rerio). Front Neuroanat 2018; 12:23. [PMID: 29636669 PMCID: PMC5881088 DOI: 10.3389/fnana.2018.00023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/12/2018] [Indexed: 01/22/2023] Open
Abstract
The Reprimo (RPRM) family is composed of highly conserved single-exon genes. The expression pattern of this gene family has been recently described during zebrafish (Danio rerio) embryogenesis, and primarily locates in the nervous system. Its most characterized member, RPRM, which duplicated to give rise rprma and rprmb in the fish lineage, is known to act as a tumor-suppressor gene in mammalian models. Here, we describe in detail the spatiotemporal expression of three rprm genes (rprma, rprmb, and rprml) within distinct anatomical structures in the developing peripheral and central nervous system. In the zebrafish, rprma mRNA is expressed in the olfactory placodes (OP) and olfactory epithelium (OE), rprmb is observed in the tectum opticum (TeO) and trigeminal ganglion (Tg), whereas rprml is found primarily in the telencephalon (Tel). At protein level, RPRM is present in a subset of cells in the OP, and neurons in the OE, TeO, hindbrain and sensory peripheral structures. Most importantly, the expression of RPRM has been conserved between teleosts and mammals. Thus, we provide a reference dataset describing the expression patterns of RPRM gene products during zebrafish and mouse development as a first step to approach the physiological role of the RPRM gene family.
Collapse
Affiliation(s)
- Karen Stanic
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alonso Quiroz
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carmen G Lemus
- Anatomy and Developmental Biology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Ignacio A Wichmann
- Advanced Center for Chronic Diseases, Santiago, Chile.,UC Center for Investigation in Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases, Santiago, Chile.,UC Center for Investigation in Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Oncología y Hematología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Santiago, Chile.,UC Center for Investigation in Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Oncología y Hematología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Miguel L Concha
- Anatomy and Developmental Biology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Julio D Amigo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|