1
|
Wang L, Guo W, Tian Y, Wang J, Xu S, Shu W, Liang H, Chen M. Carboxypeptidase inhibitor Latexin (LXN) regulates intestinal organogenesis and intestinal remodeling involved in intestinal injury repair in mice. Int J Biol Macromol 2024; 279:135129. [PMID: 39208900 DOI: 10.1016/j.ijbiomac.2024.135129] [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: 03/03/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The self-renewal and regeneration of intestinal epithelium are mainly driven by intestinal stem cells resided in crypts, which are crucial for rapid recovery intestinal tissue following injury. Latexin (LXN) is a highly expressed stem cell proliferation and differentiation related gene in intestinal tissue. However, it is still ambiguous whether LXN participates in intestine regeneration by regulating intestinal stem cells (ISCs). Here, we report that LXN colocalizes with Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) in intestinal crypts, and deletion of LXN upregulates the expression of Lgr5 in intestinal crypts. LXN deficiency promotes the proliferation of ISCs, thereby enhances the development of intestinal organoids. Mechanically, we show that LXN deficiency enhances the expression of Lgr5 in ISCs by activating the Yes-associated protein (YAP) and wingless (Wnt) signal pathways, thus accelerating intestinal normal growth and regeneration post-injury. In summary, these findings uncover a novel function of LXN in intestinal regeneration post-injury and intestinal organogenesis, suggesting the potential role of LXN in the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Lingzhu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Wenwen Guo
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Yang Tian
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Jingzhu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Shaohua Xu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Biotechnology, Guilin Medical University, Guilin, China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.
| | - Ming Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Laboratory Animal Center, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.
| |
Collapse
|
2
|
He G, Ni Y, Hua R, Wan H, Tan Y, Chen Q, Xu S, Yang Y, Zhang L, Shu W, Huang KB, Mo Y, Liang H, Chen M. Latexin deficiency limits foam cell formation and ameliorates atherosclerosis by promoting macrophage phenotype differentiation. Cell Death Dis 2024; 15:754. [PMID: 39424784 PMCID: PMC11492231 DOI: 10.1038/s41419-024-07141-3] [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: 04/05/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Latexin (LXN) is abundant in macrophages and plays critical roles in inflammation. Much is known about macrophages in atherosclerosis, the role of macrophage LXN in atherosclerosis has remained elusive. Here, the expression of LXN in human and mouse atherosclerotic lesions was examined by immunofluorescence and immunohistochemistry. LXN knockout and LXN/ApoE double-knockout mice were generated to evaluate the functions of LXN in atherosclerosis. Bone marrow transplantation (BMT) experimentation was carried out to determine whether macrophage LXN regulates atherosclerosis. We found that LXN is enriched in human and murine atherosclerotic lesions, mainly localized to macrophages. LXN deletion ameliorated atherosclerosis in ApoE-/- mice. BMT demonstrate that deletion of LXN in bone marrow protects ApoE-/- mice against atherosclerosis. Mechanistically, we found that LXN targets and inhibits JAK1 in macrophages. LXN deficiency stimulates the JAK1/STAT3/ABC transporter pathway, thereby enhancing the anti-inflammatory and anti-oxidant phenotype, cholesterol efflux, subsequently minimizing foam cell formation and atherosclerosis. Gene therapy by treatment of atherosclerotic mice with adeno-associated virus harbouring LXN-depleting shRNA attenuated the disease phenotype. In summary, our study provides new clues for the role of LXN in the pathological regulation of atherosclerosis, and determines that LXN is a target for preventing atherosclerosis, which may be a potential new anti-atherosclerosis therapeutic target.
Collapse
Affiliation(s)
- Guozhang He
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuanting Ni
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Rong Hua
- Department of Scientific Research, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huaibin Wan
- Heyuan Research Center for Cardiovascular Diseases, Department of Cardiology, the Fifth Affiliated Hospital of Jinan University, Heyuan, Guangdong, China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Qiwei Chen
- Department of Scientific Research, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shaohua Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuzhong Yang
- Department of Pathology, Affiliated Hospital of Guilin Medical College, Guilin, China
| | - Lijun Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yi Mo
- Biobank department, The reproductive hospital of Guangxi Zhuang autonomous region, Nanning, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| |
Collapse
|
3
|
Kurkowiak M, Fletcher S, Daniels A, Mozolewski P, Silvestris DA, Król E, Marek-Trzonkowska N, Hupp T, Tait-Burkard C. Differential RNA editing landscapes in host cell versus the SARS-CoV-2 genome. iScience 2023; 26:108031. [PMID: 37876814 PMCID: PMC10590966 DOI: 10.1016/j.isci.2023.108031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/09/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
The SARS-CoV-2 pandemic was defined by the emergence of new variants formed through virus mutation originating from random errors not corrected by viral proofreading and/or the host antiviral response introducing mutations into the viral genome. While sequencing information hints at cellular RNA editing pathways playing a role in viral evolution, here, we use an in vitro human cell infection model to assess RNA mutation types in two SARS-CoV-2 strains representing the original and the alpha variants. The variants showed both different cellular responses and mutation patterns with alpha showing higher mutation frequency with most substitutions observed being C-U, indicating an important role for apolipoprotein B mRNA editing catalytic polypeptide-like editing. Knockdown of select APOBEC3s through RNAi increased virus production in the original virus, but not in alpha. Overall, these data suggest a deaminase-independent anti-viral function of APOBECs in SARS-CoV-2 while the C-U editing itself might function to enhance genetic diversity enabling evolutionary adaptation.
Collapse
Affiliation(s)
- Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Sarah Fletcher
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Alison Daniels
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
- Infection Medicine, University of Edinburgh, Little France Crescent, UK
| | - Paweł Mozolewski
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | | | - Ewelina Król
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine Medical University of Gdańsk, Gdańsk, Poland
| | - Ted Hupp
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Cell Signalling Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Christine Tait-Burkard
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| |
Collapse
|
4
|
Cui X, Zhang C, Wang F, Zhao X, Wang S, Liu J, He D, Wang C, Yang FC, Tong S, Liang Y. Latexin regulates sex dimorphism in hematopoiesis via gender-specific differential expression of microRNA 98-3p and thrombospondin 1. Cell Rep 2023; 42:112274. [PMID: 36933218 PMCID: PMC10160986 DOI: 10.1016/j.celrep.2023.112274] [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: 05/19/2022] [Revised: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Hematopoietic stem cells (HSCs) have the ability to self-renew and differentiate to all blood cell types. HSCs and their differentiated progeny show sex/gender differences. The fundamental mechanisms remain largely unexplored. We previously reported that latexin (Lxn) deletion increased HSC survival and repopulation capacity in female mice. Here, we find no differences in HSC function and hematopoiesis in Lxn knockout (Lxn-/-) male mice under physiologic and myelosuppressive conditions. We further find that Thbs1, a downstream target gene of Lxn in female HSCs, is repressed in male HSCs. Male-specific high expression of microRNA 98-3p (miR98-3p) contributes to Thbs1 suppression in male HSCs, thus abrogating the functional effect of Lxn in male HSCs and hematopoiesis. These findings uncover a regulatory mechanism involving a sex-chromosome-related microRNA and its differential control of Lxn-Thbs1 signaling in hematopoiesis and shed light on the process underlying sex dimorphism in both normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Xiaojing Cui
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536 USA
| | - Cuiping Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536 USA
| | - Fang Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536 USA
| | - Xinghui Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536 USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536 USA
| | - Jinpeng Liu
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536 USA
| | - Daheng He
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536 USA
| | - Chi Wang
- Division of Cancer Biostatistics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536 USA
| | - Feng-Chun Yang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Sheng Tong
- Department of Bioengineering, University of Kentucky, Lexington, KY 40536, USA
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536 USA.
| |
Collapse
|
5
|
Li Y, Tan Y, Li X, Chen X, Wang L, Zhang L, Xu S, Huang K, Shu W, Liang H, Chen M. Loss of LXN promotes macrophage M2 polarization and PD-L2 expression contributing cancer immune-escape in mice. Cell Death Dis 2022; 8:440. [PMID: 36323670 PMCID: PMC9630456 DOI: 10.1038/s41420-022-01227-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Latexin (LXN) plays an important role in tumorigenesis and inflammatory response and as a tumor suppressor in many tumors. However, whether LXN regulates tumorigenesis through immune regulation remains uncertain. Here, we demonstrate that LXN deficiency increases hematopoietic stem cells, as well as affects the proportion of immune cells in the peripheral system. Animal studies show that mice loss of LXN promotes tumor growth in subcutaneous tumor model and AOM/DSS-induced colorectal cancer model. We found that loss of LXN promotes macrophage M2 polarization and PD-L2 expression in macrophage, thus, inhibits the function of T cells. Adoptive transfer of wild-type macrophage rescues the function of T cells in LXN-deficient mice. LXN deficiency in hematopoietic lineage exacerbates colorectal carcinogenesis, and targeted inhibition of PD-L2 ameliorates cancer growth in LXN-deficient mice. Mechanistically, we demonstrate that LXN inhibits STAT3 transcriptional activity by targeting inhibition of JAK1 in macrophages. LXN deficiency enhances PD-L2 expression rather than PD-L1 in macrophages, which lead to inhibition of T cells in tumor microenvironment. Collectively, we define a critical role of LXN/JAK1/STAT3 signal in macrophage and highlights the potential role of LXN in tumor immune-escape by regulating macrophage polarization, as well as the expression of immune checkpoint PD-L2.
Collapse
Affiliation(s)
- Yaping Li
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Yanhui Tan
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - XiuZhen Li
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Xuanming Chen
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Lingzhu Wang
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Lijun Zhang
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Shaohua Xu
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Kebing Huang
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Wei Shu
- grid.443385.d0000 0004 1798 9548College of Biotechnology, Guilin Medical University, Guilin, 541199 P.R. China
| | - Hong Liang
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| | - Ming Chen
- grid.459584.10000 0001 2196 0260State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004 P.R. China
| |
Collapse
|
6
|
Kan S, Li R, Tan Y, Yang F, Xu S, Wang L, Zhang L, Sun X, Chen X, Yang Y, Shu W, Wan H, Chen ZF, Liang H, Chen M. Latexin deficiency attenuates adipocyte differentiation and protects mice against obesity and metabolic disorders induced by high-fat diet. Cell Death Dis 2022; 13:175. [PMID: 35210404 PMCID: PMC8873487 DOI: 10.1038/s41419-022-04636-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022]
Abstract
AbstractObesity is a risk factor for many chronic diseases, and is associated with increased incidence rate of type 2 diabetes, hypertension, dyslipidemia and cardiovascular diseases. Adipocyte differentiation play critical role during development of obesity. Latexin (LXN), a mammalian carboxypeptidase inhibitor, plays important role in the proliferation and differentiation of stem cells, and highlights as a differentiation-associated gene that was significantly downregulated in prostate stem cells and whose expression increases through differentiation. However, it is unclear whether LXN is involved in adipocyte differentiation. The aim of this study was to evaluate the role of LXN on adipocyte differentiation, as well as its effects on high fat-induced obesity and metabolic disorders. In this study, we determine the expression of LXN in adipose tissue of lean and fat mice by Western blot, qPCR and immunohistochemistry. We found that LXN in fat tissues was continuous increased during the development of diet-induced obesity. We fed wild-type (WT) and LXN−/−mice with high-fat diet (HFD) to study the effects of LXN on obesity and related metabolic functions. We found that mice deficient in LXN showed resistance against high-fat diet (HFD)-induced obesity, glucose tolerance, insulin tolerance and hepatic steatosis. In vitro studies indicated that LXN was highly induced during adipocyte differentiation, and positively regulated adipocyte differentiation and adipogenesis in 3T3-L1 cells and primary preadipocytes. Functional analysis revealed that the expression of LXN was positively regulated by mTOR/RXR/PPARɤ signaling pathway during the differentiation of adipocytes, while LXN deletion decreased the protein level of PPARɤ in adipocyte through enhancing FABP4 mediated ubiquitination, which led to impaired adipocyte differentiation and lipogenesis. Collectively, our data provide evidence that LXN is a key positive regulator of adipocyte differentiation, and therapeutics targeting LXN could be effective in preventing obesity and its associated disorders in clinical settings.
Collapse
|
7
|
Wang CH, Wang LK, Wu CC, Chen ML, Kuo CY, Shyu RY, Tsai FM. Cathepsin V Mediates the Tazarotene-induced Gene 1-induced Reduction in Invasion in Colorectal Cancer Cells. Cell Biochem Biophys 2020; 78:483-494. [PMID: 32918681 DOI: 10.1007/s12013-020-00940-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) is a retinoid acid receptor-responsive gene involved in cell differentiation and tumorigenesis. Aberrant methylation of CpG islands in the TIG1 promoter is found in multiple cancers. Currently, the exact mechanism underlying the anticancer effect of TIG1 is unknown. Here, we show that TIG1 interacts with cathepsin V (CTSV), which reduces CTSV stability and subsequently affects the production of activated urokinase-type plasminogen activator (uPA), an epithelial-mesenchymal transition-associated protein. Ectopic expression of CTSV increased the expression of activated uPA and the number of migrated and invaded cells, whereas ectopic TIG1 expression reversed the effects of CTSV on the uPA signaling pathway. Similar patterns in the production of activated uPA and number of migrated and invaded cells were also observed in TIG1-expressing and CTSV-knockdown cells. The results suggest that CTSV may participate in TIG1-regulated uPA activity and the associated downstream signaling pathway.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Dermatology, Taipei Tzuchi Hospital, Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou, Taoyuan, 333, Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, 202, Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| |
Collapse
|
8
|
Latexin deficiency in mice up-regulates inflammation and aggravates colitis through HECTD1/Rps3/NF-κB pathway. Sci Rep 2020; 10:9868. [PMID: 32555320 PMCID: PMC7299958 DOI: 10.1038/s41598-020-66789-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
The function of Latexin (LXN) in inflammation has attracted attention. However, no data are available regarding its role in colitis. We report that LXN is a suppressor of colitis. LXN deficiency leads to the severity of colitis in DSS-induced mice, and LXN is required for the therapeutic effect of retinoic acid on colitis. Using a proteomics approach, we demonstrate that LXN interacts and forms a functional complex with HECTD1 (an E3 ubiquitin ligase) and ribosomal protein subunit3 (Rps3). IκBα is one of the substrates of HECTD1. Ectopic expression of LXN leads to IκBα accumulation in intestinal epithelial cells, however, LXN knockdown enhances the interaction of HECTD1 and Rps3, contributing to the ubiquitination degradation of IκBα, and subsequently enhances inflammatory response. Thus, our findings provided a novel mechanism underlying LXN modulates colitis via HECTD1/Rps3/NF-κB pathway and significant implications for the development of novel strategies for the treatment of colitis by targeting LXN.
Collapse
|
9
|
Morishima T, Takizawa H. Genetic fingerprint defines hematopoietic stem cell pool size and function. Haematologica 2020; 105:526-528. [PMID: 32115410 DOI: 10.3324/haematol.2019.241299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Tatsuya Morishima
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University .,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences, Kumamoto University
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University .,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
Liu Y, Dou M, Song X, Dong Y, Liu S, Liu H, Tao J, Li W, Yin X, Xu W. The emerging role of the piRNA/piwi complex in cancer. Mol Cancer 2019; 18:123. [PMID: 31399034 PMCID: PMC6688334 DOI: 10.1186/s12943-019-1052-9] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/31/2019] [Indexed: 01/08/2023] Open
Abstract
Piwi interacting RNAs (piRNAs) constitute novel small non-coding RNA molecules of approximately 24-31 nucleotides in length that often bind to members of the piwi protein family to play regulatory roles. Recently, emerging evidence suggests that in addition to the mammalian germline, piRNAs are also expressed in a tissue-specific manner in a variety of human tissues and modulate key signaling pathways at the transcriptional or post-transcriptional level. In addition, a growing number of studies have shown that piRNA and PIWI proteins, which are abnormally expressed in various cancers, may serve as novel biomarkers and therapeutic targets for tumor diagnostics and treatment. However, the functions of piRNAs in cancer and their underlying mechanisms remain incompletely understood. In this review, we discuss current findings regarding piRNA biogenetic processes, functions, and emerging roles in cancer, providing new insights regarding the potential applications of piRNAs and piwi proteins in cancer diagnosis and clinical treatment.
Collapse
Affiliation(s)
- Yongmei Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Mei Dou
- School of Public Health, Qingdao University, Qingdao, 266003, China
| | - Xuxia Song
- The Laboratory of Biomedical Center, Qingdao University, Qingdao, 266003, China
| | - Yanhan Dong
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Si Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Haoran Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Jiaping Tao
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Wenjing Li
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Xunhua Yin
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China
| | - Wenhua Xu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
11
|
Zhang C, Fondufe-Mittendorf YN, Wang C, Chen J, Cheng Q, Zhou D, Zheng Y, Geiger H, Liang Y. Latexin regulation by HMGB2 is required for hematopoietic stem cell maintenance. Haematologica 2019; 105:573-584. [PMID: 31171637 PMCID: PMC7049332 DOI: 10.3324/haematol.2018.207092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 06/05/2019] [Indexed: 01/12/2023] Open
Abstract
Hematopoietic stem cells provide life-long production of blood cells and undergo self-renewal division in order to sustain the stem cell pool. Homeostatic maintenance of hematopoietic stem cell pool and blood cell production is vital for the organism to survive. We previously reported that latexin is a negative regulator of hematopoietic stem cells in mice. Its natural variation in the expression is inversely correlated with hematopoietic stem cell number. However, the molecular mechanisms regulating latexin transcription remain largely unknown, and the genetic factors contributing to its natural variation are not clearly defined. Here we discovered a chromatin protein, high-mobility group protein B2, as a novel transcriptional suppressor of latexin by using DNA pull-down and mass spectrometry. High-mobility group protein B2 knockdown increases latexin expression at transcript and protein levels, and decreases hematopoietic stem cell number and regeneration capacity in vivo. Concomitant blockage of latexin activation significantly reverses these phenotypic changes, suggesting that latexin is one of the downstream targets and functional mediators of high-mobility group protein B2. We further identified a functional single nucleotide polymorphism, rs31528793, in the latexin promoter that binds to high-mobility group protein B2 and affects the promoter activity. G allelic variation in rs31528793 associates with the higher latexin expression and lower hematopoietic stem cell number, whereas C allele indicates the lower latexin expression and higher stem cell number. This study reveals for the first time that latexin transcription is regulated by both transacting (high-mobility group protein B2) and cis-acting (single nucleotide polymorphism rs31528793) factors. It uncovers the functional role of naturally occurring genetic variants, in combination with epigenetic regulator, in determining differential gene expression and phenotypic diversity in the hematopoietic stem cell population.
Collapse
Affiliation(s)
- Cuiping Zhang
- Departments of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | | | - Chi Wang
- Department of Cancer Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Jin Chen
- Department of Internal Medicine and Computer Science, University of Kentucky, Lexington, KY, USA
| | - Qiang Cheng
- Department of Internal Medicine and Computer Science, University of Kentucky, Lexington, KY, USA
| | - Daohong Zhou
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Yi Zheng
- Cincinnati Children's Hospital Medical Center, Experimental Hematology and Cancer Biology, Cincinnati, OH, USA
| | - Hartmut Geiger
- Cincinnati Children's Hospital Medical Center, Experimental Hematology and Cancer Biology, Cincinnati, OH, USA.,Institute for Molecular Medicine, University of Ulm, Ulm, Germany
| | - Ying Liang
- Departments of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
12
|
The putative tumour suppressor protein Latexin is secreted by prostate luminal cells and is downregulated in malignancy. Sci Rep 2019; 9:5120. [PMID: 30914656 PMCID: PMC6435711 DOI: 10.1038/s41598-019-41379-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/28/2019] [Indexed: 11/09/2022] Open
Abstract
Loss of latexin (LXN) expression negatively correlates with the prognosis of several human cancers. Despite association with numerous processes including haematopoietic stem cell (HSC) fate, inflammation and tumour suppression, a clearly defined biological role for LXN is still lacking. Therefore, we sought to understand LXN expression and function in the normal and malignant prostate to assess its potential as a therapeutic target. Our data demonstrate that LXN is highly expressed in normal prostate luminal cells but downregulated in high Gleason grade cancers. LXN protein is both cytosolic and secreted by prostate cells and expression is directly and potently upregulated by all-trans retinoic acid (atRA). Whilst overexpression of LXN in prostate epithelial basal cells did not affect cell fate, LXN overexpression in the luminal cancer line LNCaP reduced plating efficiency. Transcriptome analysis revealed that LXN overexpression had no direct effects on gene expression but had significant indirect effects on important genes involved in both retinoid metabolism and IFN-associated inflammatory responses. These data highlight a potential role for LXN in retinoid signaling and inflammatory pathways. Investigating the effects of LXN on immune cell function in the tumour microenvironment (TME) may reveal how observed intratumoural loss of LXN affects the prognosis of many adenocarcinomas.
Collapse
|
13
|
Maimouni S, Lee MH, Sung YM, Hall M, Roy A, Ouaari C, Hwang YS, Spivak J, Glasgow E, Swift M, Patel J, Cheema A, Kumar D, Byers S. Tumor suppressor RARRES1 links tubulin deglutamylation to mitochondrial metabolism and cell survival. Oncotarget 2019; 10:1606-1624. [PMID: 30899431 PMCID: PMC6422194 DOI: 10.18632/oncotarget.26600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
RARRES1, a retinoic acid regulated carboxypeptidase inhibitor associated with fatty acid metabolism, stem cell differentiation and tumorigenesis is among the most commonly methylated loci in multiple cancers but has no known mechanism of action. Here we show that RARRES1 interaction with cytoplasmic carboxypeptidase 2 (CCP2) inhibits tubulin deglutamylation, which in turn regulates the mitochondrial voltage dependent anion channel (VDAC1), mitochondrial membrane potential, AMPK activation, energy balance and metabolically reprograms cells and zebrafish to a more energetic and anabolic phenotype. Depletion of RARRES1 also increases expression of stem cell markers, promotes anoikis, anchorage independent growth and insensitivity to multiple apoptotic stimuli. As depletion of CCP2 or inhibition of VDAC1 reverses the effects of RARRES1 depletion on energy balance and cell survival we conclude that RARRES1 modulation of CCP2-modulated tubulin-mitochondrial VDAC1 interactions is a fundamental regulator of cancer and stem cell metabolism and survival.
Collapse
Affiliation(s)
- Sara Maimouni
- Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| | - Mi-Hye Lee
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - You-Me Sung
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Michael Hall
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Arpita Roy
- University of the District of Columbia, Washington, DC, USA
| | - Chokri Ouaari
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.,University of the District of Columbia, Washington, DC, USA
| | - Yoo-Seok Hwang
- Cancer & Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, MD, USA
| | - Justin Spivak
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eric Glasgow
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Matthew Swift
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Jay Patel
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Amrita Cheema
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Deepak Kumar
- University of the District of Columbia, Washington, DC, USA
| | - Stephen Byers
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.,Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) produce mature blood cells throughout lifetime. Natural genetic diversity offers an important yet largely untapped reservoir for deciphering regulatory mechanisms of HSCs and hematopoiesis. In this review, we explore the role of latexin, identified by natural variation, in regulating homeostatic and stress hematopoiesis, unravel the underlying signaling pathways, and propose its therapeutic implication. RECENT FINDINGS Latexin acts endogenously in HSCs to negatively regulate their population size by enhancing apoptosis and by decreasing self-renewal. Deletion of latexin in vivo increases HSC repopulation capacity and survival, expands the entire hematopoietic system, and mitigates myelosuppression. Latexin inactivation downregulates thrombospondin 1 (Thbs1). It inhibits nuclear translocation of ribosomal protein subunit 3 (Rps3), a novel latexin-binding protein, and sensitizes hematopoietic cells to radiation-induced cell death. However, how latexin-Rps3 pathway regulates Thbs1 transcription is unclear. Latexin is downregulated in cancer cells because of promoter hypermethylation, but latexin-depleted mice do not inherently develop hematologic malignancies even with aging. The mechanism of action of latexin in tumorigenesis remains largely unknown. SUMMARY Understanding how latexin regulates HSC survival, self-renewal, and stress response will advance our knowledge of HSC biology. It will facilitate the development of a novel therapeutic strategy for hematopoietic regeneration and cancer treatment.
Collapse
|
15
|
Gerber T, Willscher E, Loeffler-Wirth H, Hopp L, Schadendorf D, Schartl M, Anderegg U, Camp G, Treutlein B, Binder H, Kunz M. Mapping heterogeneity in patient-derived melanoma cultures by single-cell RNA-seq. Oncotarget 2018; 8:846-862. [PMID: 27903987 PMCID: PMC5352202 DOI: 10.18632/oncotarget.13666] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/12/2016] [Indexed: 01/21/2023] Open
Abstract
Recent technological advances in single-cell genomics make it possible to analyze cellular heterogeneity of tumor samples. Here, we applied single-cell RNA-seq to measure the transcriptomes of 307 single cells cultured from three biopsies of three different patients with a BRAF/NRAS wild type, BRAF mutant/NRAS wild type and BRAF wild type/NRAS mutant melanoma metastasis, respectively. Analysis based on self-organizing maps identified sub-populations defined by multiple gene expression modules involved in proliferation, oxidative phosphorylation, pigmentation and cellular stroma. Gene expression modules had prognostic relevance when compared with gene expression data from published melanoma samples and patient survival data. We surveyed kinome expression patterns across sub-populations of the BRAF/NRAS wild type sample and found that CDK4 and CDK2 were consistently highly expressed in the majority of cells, suggesting that these kinases might be involved in melanoma progression. Treatment of cells with the CDK4 inhibitor palbociclib restricted cell proliferation to a similar, and in some cases greater, extent than MAPK inhibitors. Finally, we identified a low abundant sub-population in this sample that highly expressed a module containing ABC transporter ABCB5, surface markers CD271 and CD133, and multiple aldehyde dehydrogenases (ALDHs). Patient-derived cultures of the BRAF mutant/NRAS wild type and BRAF wild type/NRAS mutant metastases showed more homogeneous single-cell gene expression patterns with gene expression modules for proliferation and ABC transporters. Taken together, our results describe an intertumor and intratumor heterogeneity in melanoma short-term cultures which might be relevant for patient survival, and suggest promising targets for new treatment approaches in melanoma therapy.
Collapse
Affiliation(s)
- Tobias Gerber
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology Leipzig, 04103 Leipzig, Germany
| | - Edith Willscher
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Lydia Hopp
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, 45147 Essen, Germany
| | - Manfred Schartl
- Department of Physiological Chemistry, University of Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, 97080 Würzburg, Germany.,Institute for Advanced Study, 3572 Texas A&M University, College Station, Texas 77843-3572, USA
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany
| | - Gray Camp
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology Leipzig, 04103 Leipzig, Germany
| | - Barbara Treutlein
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology Leipzig, 04103 Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
16
|
The effect of calorie restriction on mouse skeletal muscle is sex, strain and time-dependent. Sci Rep 2017; 7:5160. [PMID: 28698572 PMCID: PMC5505993 DOI: 10.1038/s41598-017-04896-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
Loss of skeletal muscle mass and function occurs with increasing age. Calorie restriction (CR) increases the lifespan of C57Bl/6 mice, but not in the shorter-lived DBA/2 strain. There is some evidence that calorie restriction reduces or delays many of the age-related defects that occur in rodent skeletal muscle. We therefore investigated the effect of short (2.5 month) and longer term (8.5 and 18.5 months) CR on skeletal muscle in male and female C57Bl/6 and DBA/2 mice. We found that short-term CR increased the satellite cell number and collagen VI content of muscle, but resulted in a delayed regenerative response to injury.Consistent with this, the in vitro proliferation of satellite cells derived from these muscles was reduced by CR. The percentage of stromal cells, macrophages, hematopoietic stem cells and fibroadipogenic cells in the mononucleated cell population derived from skeletal muscle was reduced by CR at various stages. But overall, these changes are neither consistent over time, nor between strain and sex. The fact that changes induced by CR do not persist with time and the dissimilarities between the two mouse strains, combined with sex differences, urge caution in applying CR to improve skeletal muscle function across the lifespan in humans.
Collapse
|
17
|
Kanatsu-Shinohara M, Tanaka T, Ogonuki N, Ogura A, Morimoto H, Cheng PF, Eisenman RN, Trumpp A, Shinohara T. Myc/Mycn-mediated glycolysis enhances mouse spermatogonial stem cell self-renewal. Genes Dev 2017; 30:2637-2648. [PMID: 28007786 PMCID: PMC5204355 DOI: 10.1101/gad.287045.116] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022]
Abstract
Here, Kanatsu-Shinohara et al. investigated the mechanisms underlying Myc regulation of spermatogonial stem cell (SSC) fate. Their findings suggest that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division. Myc plays critical roles in the self-renewal division of various stem cell types. In spermatogonial stem cells (SSCs), Myc controls SSC fate decisions because Myc overexpression induces enhanced self-renewal division, while depletion of Max, a Myc-binding partner, leads to meiotic induction. However, the mechanism by which Myc acts on SSC fate is unclear. Here we demonstrate a critical link between Myc/Mycn gene activity and glycolysis in SSC self-renewal. In SSCs, Myc/Mycn are regulated by Foxo1, whose deficiency impairs SSC self-renewal. Myc/Mycn-deficient SSCs not only undergo limited self-renewal division but also display diminished glycolytic activity. While inhibition of glycolysis decreased SSC activity, chemical stimulation of glycolysis or transfection of active Akt1 or Pdpk1 (phosphoinositide-dependent protein kinase 1 ) augmented self-renewal division, and long-term SSC cultures were derived from a nonpermissive strain that showed limited self-renewal division. These results suggested that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Kyoto 606-8501, Japan
| | - Takashi Tanaka
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | | | - Atsuo Ogura
- Bioresource Center, RIKEN, Tsukuba 305-0074, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Pei Feng Cheng
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforshungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
18
|
Liu Y, Zhang C, Li Z, Wang C, Jia J, Gao T, Hildebrandt G, Zhou D, Bondada S, Ji P, St Clair D, Liu J, Zhan C, Geiger H, Wang S, Liang Y. Latexin Inactivation Enhances Survival and Long-Term Engraftment of Hematopoietic Stem Cells and Expands the Entire Hematopoietic System in Mice. Stem Cell Reports 2017; 8:991-1004. [PMID: 28330618 PMCID: PMC5390104 DOI: 10.1016/j.stemcr.2017.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/20/2022] Open
Abstract
Natural genetic diversity offers an important yet largely untapped resource to decipher the molecular mechanisms regulating hematopoietic stem cell (HSC) function. Latexin (Lxn) is a negative stem cell regulatory gene identified on the basis of genetic diversity. By using an Lxn knockout mouse model, we found that Lxn inactivation in vivo led to the physiological expansion of the entire hematopoietic hierarchy. Loss of Lxn enhanced the competitive repopulation capacity and survival of HSCs in a cell-intrinsic manner. Gene profiling of Lxn-null HSCs showed altered expression of genes enriched in cell-matrix and cell-cell interactions. Thrombospondin 1 (Thbs1) was a potential downstream target with a dramatic downregulation in Lxn-null HSCs. Enforced expression of Thbs1 restored the Lxn inactivation-mediated HSC phenotypes. This study reveals that Lxn plays an important role in the maintenance of homeostatic hematopoiesis, and it may lead to development of safe and effective approaches to manipulate HSCs for clinical benefit.
Collapse
Affiliation(s)
- Yi Liu
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Cuiping Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA
| | - Zhenyu Li
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Department of Cancer Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Jianhang Jia
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Tianyan Gao
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Gerhard Hildebrandt
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Peng Ji
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| | - Daret St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA
| | - Jinze Liu
- Department of Computer Science, University of Kentucky, Lexington, KY 40536, USA
| | - Changguo Zhan
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Hartmut Geiger
- Cincinnati Children's Hospital Medical Center, Experimental Hematology and Cancer Biology, Cincinnati, OH 45229, USA; Institute for Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Health Sciences Research Building Room 340, 1095 V.A. Drive, Lexington, KY 40536, USA.
| |
Collapse
|
19
|
The accumulation of lipids and proteins during red blood cell storage: the roles of leucoreduction and experimental filtration. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2017; 15:131-136. [PMID: 28263170 DOI: 10.2450/2017.0314-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023]
Abstract
Pre-storage leucoreduction has been universally adopted in most developed countries in Asia, Europe and the Americas. It decreases febrile transfusion reactions, alloimmunisation to HLA antigens, cytomegalovirus exposure, the accumulation of a number of pro-inflammatory mediators in the supernatant, including the accumulation of platelet-and leucocyte-derived proteins and metabolites during routine storage. This review will highlight the lipids and proteins, biological response modifiers (BRMs) that accumulate, their clinical effects in transfused hosts, and methods of mitigation.
Collapse
|
20
|
Zhao Y, Carroll DW, You Y, Chaiswing L, Wen R, Batinic-Haberle I, Bondada S, Liang Y, St Clair DK. A novel redox regulator, MnTnBuOE-2-PyP 5+, enhances normal hematopoietic stem/progenitor cell function. Redox Biol 2017; 12:129-138. [PMID: 28231483 PMCID: PMC5320058 DOI: 10.1016/j.redox.2017.02.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 12/22/2022] Open
Abstract
The signaling of reactive oxygen species (ROS) is essential for the maintenance of normal cellular function. However, whether and how ROS regulate stem cells are unclear. Here, we demonstrate that, in transgenic mice expressing the human manganese superoxide dismutase (MnSOD) gene, a scavenger of ROS in mitochondria, the number and function of mouse hematopoietic stem/progenitor cells (HSPC) under physiological conditions are enhanced. Importantly, giving MnTnBuOE-2-PyP5+(MnP), a redox- active MnSOD mimetic, to mouse primary bone marrow cells or to C57B/L6 mice significantly enhances the number of HSPCs. Mechanistically, MnP reduces superoxide to hydrogen peroxide, which activates intracellular Nrf2 signaling leading to the induction of antioxidant enzymes, including MnSOD and catalase, and mitochondrial uncoupling protein 3. The results reveal a novel role of ROS signaling in regulating stem cell function, and suggest a possible beneficial effect of MnP in treating pathological bone marrow cell loss and in increasing stem cell population for bone marrow transplantation.
Collapse
Affiliation(s)
- Y Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - D W Carroll
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Y You
- Department of Neurosurgery, University of Texas, Houston, TX, USA
| | - L Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - R Wen
- Genetic Center, Women and Children's Healthcare, Qingdao, China
| | - I Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - S Bondada
- Department of Microbiology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Y Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - D K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
21
|
Zhang M, Osisami M, Dai J, Keller JM, Escara-Wilke J, Mizokami A, Keller ET. Bone Microenvironment Changes in Latexin Expression Promote Chemoresistance. Mol Cancer Res 2017; 15:457-466. [PMID: 28087740 DOI: 10.1158/1541-7786.mcr-16-0392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
Although docetaxel is the standard of care for advanced prostate cancer, most patients develop resistance to docetaxel. Therefore, elucidating the mechanism that underlies resistance to docetaxel is critical to enhance therapeutic intervention. Mining cDNA microarray from the PC-3 prostate cancer cell line and its docetaxel-resistant derivative (PC3-TxR) revealed decreased latexin (LXN) expression in the resistant cells. LXN expression was inversely correlated with taxane resistance in a panel of prostate cancer cell lines. LXN knockdown conferred docetaxel resistance to prostate cancer cells in vitro and in vivo, whereas LXN overexpression reduced docetaxel resistance in several prostate cancer cell lines. A mouse model of prostate cancer demonstrated that prostate cancer cells developed resistance to docetaxel in the bone microenvironment, but not the soft tissue microenvironment. This was associated with decreased LXN expression in prostate cancer cells in the bone microenvironment compared with the soft tissue microenvironment. It was identified that bone stromal cells decreased LXN expression through methylation and induced chemoresistance in prostate cancer in vitro These findings reveal that a subset of prostate cancer develops docetaxel resistance through loss of LXN expression associated with methylation and that the bone microenvironment promotes this drug resistance phenotype.Implications: This study suggests that the LXN pathway should be further explored as a viable target for preventing or reversing taxane resistance in prostate cancer. Mol Cancer Res; 15(4); 457-66. ©2017 AACR.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Urology, University of Michigan, Ann Arbor, Michigan.,Clinical Medicine Program, Xiangya Hospital, Central South University, Changsha, China
| | - Mary Osisami
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Jinlu Dai
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Jill M Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan.,Unit for Laboratory Animal Medicine, University of Michigan; Ann Arbor, Michigan
| | | | | | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan. .,Biointerfaces Institute, University of Michigan; Ann Arbor, Michigan
| |
Collapse
|
22
|
Shyu RY, Wang CH, Wu CC, Chen ML, Lee MC, Wang LK, Jiang SY, Tsai FM. Tazarotene-Induced Gene 1 Enhanced Cervical Cell Autophagy through Transmembrane Protein 192. Mol Cells 2016; 39:877-887. [PMID: 27989102 PMCID: PMC5223105 DOI: 10.14348/molcells.2016.0161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 01/18/2023] Open
Abstract
Tazarotene-induced gene 1 (TIG1) is a retinoic acid-inducible protein that is considered a putative tumor suppressor. The expression of TIG1 is decreased in malignant prostate carcinoma or poorly differentiated colorectal adenocarcinoma, but TIG1 is present in benign or well-differentiated tumors. Ectopic TIG1 expression led to suppression of growth in cancer cells. However, the function of TIG1 in cell differentiation is still unknown. Using a yeast two-hybrid system, we found that transmembrane protein 192 (TMEM192) interacted with TIG1. We also found that both TIG1A and TIG1B isoforms interacted and co-localized with TMEM192 in HtTA cervical cancer cells. The expression of TIG1 induced the expression of autophagy-related proteins, including Beclin-1 and LC-3B. The silencing of TMEM192 reduced the TIG1-mediated upregulation of autophagic activity. Furthermore, silencing of either TIG1 or TMEM192 led to alleviation of the upregulation of autophagy induced by all-trans retinoic acid. Our results demonstrate that the expression of TIG1 leads to cell autophagy through TMEM192. Our study also suggests that TIG1 and TMEM192 play an important role in the all-trans retinoic acid-mediated upregulation of autophagic activity.
Collapse
Affiliation(s)
- Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| | - Chun-Hua Wang
- Department of Dermatology, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114,
Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| | - Ming-Cheng Lee
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou, Taoyuan 333,
Taiwan
| | - Shun-Yuan Jiang
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City 231,
Taiwan
| |
Collapse
|
23
|
Quantitative trait gene Slit2 positively regulates murine hematopoietic stem cell numbers. Sci Rep 2016; 6:31412. [PMID: 27503415 PMCID: PMC4977545 DOI: 10.1038/srep31412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 07/21/2016] [Indexed: 12/30/2022] Open
Abstract
Hematopoietic stem cells (HSC) demonstrate natural variation in number and function. The genetic factors responsible for the variations (or quantitative traits) are largely unknown. We previously identified a gene whose differential expression underlies the natural variation of HSC numbers in C57BL/6 (B6) and DBA/2 (D2) mice. We now report the finding of another gene, Slit2, on chromosome 5 that also accounts for variation in HSC number. In reciprocal chromosome 5 congenic mice, introgressed D2 alleles increased HSC numbers, whereas B6 alleles had the opposite effect. Using gene array and quantitative polymerase chain reaction, we identified Slit2 as a quantitative trait gene whose expression was positively correlated with the number of HSCs. Ectopic expression of Slit2 not only increased the number of the long-term colony forming HSCs, but also enhanced their repopulation capacity upon transplantation. Therefore, Slit2 is a novel quantitative trait gene and a positive regulator of the number and function of murine HSCs. This finding suggests that Slit2 may be a potential therapeutic target for the effective in vitro and in vivo expansion of HSCs without compromising normal hematopoiesis.
Collapse
|
24
|
Cuvertino S, Lacaud G, Kouskoff V. SOX7-enforced expression promotes the expansion of adult blood progenitors and blocks B-cell development. Open Biol 2016; 6:160070. [PMID: 27411892 PMCID: PMC4967825 DOI: 10.1098/rsob.160070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/22/2016] [Indexed: 12/29/2022] Open
Abstract
During embryogenesis, the three SOXF transcription factors, SOX7, SOX17 and SOX18, regulate the specification of the cardiovascular system and are also involved in the development of haematopoiesis. The ectopic expression of SOX17 in both embryonic and adult blood cells enhances self-renewal. Likewise, the enforced expression of SOX7 during embryonic development promotes the proliferation of early blood progenitors and blocks lineage commitment. However, whether SOX7 expression can also affect the self-renewal of adult blood progenitors has never been explored. In this study, we demonstrate using an inducible transgenic mouse model that the enforced expression of Sox7 ex vivo in bone marrow/stroma cell co-culture promotes the proliferation of blood progenitors which retain multi-lineage short-term engrafting capacity. Furthermore, SOX7 expression induces a profound block in the generation of B lymphocytes. Correspondingly, the ectopic expression of SOX7 in vivo results in dramatic alterations of the haematopoietic system, inducing the proliferation of blood progenitors in the bone marrow while blocking B lymphopoiesis. In addition, SOX7 expression induces extra-medullary haematopoiesis in the spleen and liver. Together, these data demonstrate that the uncontrolled expression of the transcription factor SOX7 in adult haematopoietic cells has dramatic consequences on blood homeostasis.
Collapse
Affiliation(s)
- Sara Cuvertino
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Valerie Kouskoff
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
25
|
Xue Z, Zhou Y, Wang C, Zheng J, Zhang P, Zhou L, Wu L, Shan Y, Ye M, He Y, Cai Z. Latexin exhibits tumor-suppressor potential in pancreatic ductal adenocarcinoma. Oncol Rep 2015; 35:50-8. [PMID: 26530530 PMCID: PMC4699618 DOI: 10.3892/or.2015.4353] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 09/16/2015] [Indexed: 12/29/2022] Open
Abstract
Recent studies suggest that latexin (Lxn) expression is involved in stem cell regulation and that it plays significant roles in tumor cell migration and invasion. The clinicopathological significance of Lxn expression and its possible correlation with CD133 expression in pancreatic ductal adenocarcinoma (PDAC) is currently unknown. In the present study, immunohistochemical analysis was performed to determine Lxn and CD133 expression in 43 PDAC patient samples and in 32 corresponding adjacent non-cancerous samples. The results were analyzed and compared with patient age, gender, tumor site and size, histological grade, clinical stage and overall mean survival time. Lxn expression was clearly decreased in the PDAC tissues compared with that in the adjacent non-cancerous tissues, while CD133 expression was increased. Low Lxn expression in the PDAC tissues was significantly correlated with tumor size (P=0.002), histological grade (P=0.000), metastasis (P=0.007) and clinical stage (P=0.018), but not with age (P=0.451), gender (P=0.395) or tumor site (P=0.697). Kaplan-Meier survival analysis revealed that low Lxn expression was significantly correlated with reduced overall survival time (P=0.000). Furthermore, Lxn expression was found to be inversely correlated with CD133 expression (r=−0.485, P=0.001). Furthermore, CD133-positive MIA PaCa-2 pancreatic tumor cells were sorted by magnetic-activated cell sorting (MACS), and those that overexpressed Lxn exhibited a significantly higher rate of apoptosis and lower proliferative activity. Our findings suggest that Lxn may function as a tumor suppressor that targets CD133-positive pancreatic cancer cells.
Collapse
Affiliation(s)
- Zhanxiong Xue
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuhui Zhou
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Cheng Wang
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jihang Zheng
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Pu Zhang
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lingling Zhou
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Liang Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yunfeng Shan
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengsi Ye
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yun He
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhenzhai Cai
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
26
|
Zhou X, Crow AL, Hartiala J, Spindler TJ, Ghazalpour A, Barsky LW, Bennett BJ, Parks BW, Eskin E, Jain R, Epstein JA, Lusis AJ, Adams GB, Allayee H. The Genetic Landscape of Hematopoietic Stem Cell Frequency in Mice. Stem Cell Reports 2015; 5:125-38. [PMID: 26050929 PMCID: PMC4618249 DOI: 10.1016/j.stemcr.2015.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 12/31/2022] Open
Abstract
Prior efforts to identify regulators of hematopoietic stem cell physiology have relied mainly on candidate gene approaches with genetically modified mice. Here we used a genome-wide association study (GWAS) strategy with the hybrid mouse diversity panel to identify the genetic determinants of hematopoietic stem/progenitor cell (HSPC) frequency. Among 108 strains, we observed ∼120- to 300-fold variation in three HSPC populations. A GWAS analysis identified several loci that were significantly associated with HSPC frequency, including a locus on chromosome 5 harboring the homeodomain-only protein gene (Hopx). Hopx previously had been implicated in cardiac development but was not known to influence HSPC biology. Analysis of the HSPC pool in Hopx−/− mice demonstrated significantly reduced cell frequencies and impaired engraftment in competitive repopulation assays, thus providing functional validation of this positional candidate gene. These results demonstrate the power of GWAS in mice to identify genetic determinants of the hematopoietic system. Genetic variation across mouse strains influences hematopoietic stem cell frequency This variation can be exploited for genome-wide association studies Hopx is a regulator of hematopoietic stem/progenitor cell function This approach can be used to identify genetic determinants of other stem cell systems
Collapse
Affiliation(s)
- Xiaoying Zhou
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amanda L Crow
- Department of Preventive Medicine and Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jaana Hartiala
- Department of Preventive Medicine and Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tassja J Spindler
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anatole Ghazalpour
- Departments of Human Genetics, Medicine, and Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Lora W Barsky
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brian J Bennett
- Department of Genetics and Nutrition Research Institute, University of North Carolina, Chapel Hill, Kannapolis, NC 28081, USA
| | - Brian W Parks
- Departments of Human Genetics, Medicine, and Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Eleazar Eskin
- Department of Computer Science and Inter-Departmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology and Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology and Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aldons J Lusis
- Departments of Human Genetics, Medicine, and Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Gregor B Adams
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Hooman Allayee
- Department of Preventive Medicine and Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
27
|
Oren Y, Nachshon A, Frishberg A, Wilentzik R, Gat-Viks I. Linking traits based on their shared molecular mechanisms. eLife 2015; 4. [PMID: 25781485 PMCID: PMC4362207 DOI: 10.7554/elife.04346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/20/2015] [Indexed: 12/29/2022] Open
Abstract
There is growing recognition that co-morbidity and co-occurrence of disease traits are often determined by shared genetic and molecular mechanisms. In most cases, however, the specific mechanisms that lead to such trait-trait relationships are yet unknown. Here we present an analysis of a broad spectrum of behavioral and physiological traits together with gene-expression measurements across genetically diverse mouse strains. We develop an unbiased methodology that constructs potentially overlapping groups of traits and resolves their underlying combination of genetic loci and molecular mechanisms. For example, our method predicts that genetic variation in the Klf7 gene may influence gene transcripts in bone marrow-derived myeloid cells, which in turn affect 17 behavioral traits following morphine injection; this predicted effect of Klf7 is consistent with an in vitro perturbation of Klf7 in bone marrow cells. Our analysis demonstrates the utility of studying hidden causative mechanisms that lead to relationships between complex traits.
Collapse
Affiliation(s)
- Yael Oren
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Aharon Nachshon
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amit Frishberg
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Roni Wilentzik
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Irit Gat-Viks
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Xue ZX, Zheng JH, Zheng ZQ, Cai JL, Ye XH, Wang C, Sun WJ, Zhou X, Lu MD, Li PH, Cai ZZ. Latexin inhibits the proliferation of CD133+ miapaca-2 pancreatic cancer stem-like cells. World J Surg Oncol 2014; 12:404. [PMID: 25551472 PMCID: PMC4396164 DOI: 10.1186/1477-7819-12-404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 12/13/2014] [Indexed: 12/18/2022] Open
Abstract
Background An increasing number of evidence suggests that pancreatic cancer contains cancer stem cells (CSCs), which may be relevant to the resistance of chemotherapy. Latexin (Lxn) is a negative regulator of stem cell proliferation and we investigate the effects of Lxn on CD133+ pancreatic cancer stem-like cells. Methods CD133+ miapaca-2 cells, a human pancreatic carcinoma cell line, were isolated and sorted by magnetic activated cell sorting and flow cytometry. The capacity for self-renewal, proliferation, and tumorigenicity of CD133+ miapaca-2 cells was determined by the floating spheres test and tumor xenograft assays. Protein and mRNA expression of Lxn in CD133+ and CD133- miapaca-2 cells were detected by Western blotting and qRT-PCR, respectively. After CD133+ miapaca-2 cells were treated with Lxn in serum-free medium (SFM), cell proliferation was assayed with a Cell Counting Kit 8 (CCK-8) and apoptosis was analyzed by flow cytometry. The protein and mRNA expression levels of Bcl-2, bax, and c-myc were also analyzed. Results We successfully isolated CD133+ miapaca-2 cells that exhibited the capacity for self-renewal in SFM, a proliferation potential in DMEM supplemented with FBS, and high tumorigenicity in nude mice. Lxn protein and mRNA expression levels in CD133+ miapaca-2 cells were significantly lower than those in CD133- cells. Lxn-treated CD133+ miapaca-2 cells exhibited increased apoptosis and low proliferation activity, down-regulation of Bcl-2 and c-myc expression, and up-regulation of Bax expression in a dose-dependent manner. Conclusions Lxn induces apoptosis and inhibits the proliferation of CD133+ miapaca-2 cells. These changes are associated with down-regulation of Bcl-2 and c-myc and up-regulation of Bax.
Collapse
Affiliation(s)
- Zhan-Xiong Xue
- Department of Gastroenterology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Ji-Hang Zheng
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Zhi-Qiang Zheng
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Jing-Li Cai
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Xiao-Hua Ye
- Departments of Gastroenterology and Hepatology, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Jinhua, 321000, Zhejiang Province, China.
| | - Cheng Wang
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Wei-Jian Sun
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Xiang Zhou
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Ming-Dong Lu
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Pi-Hong Li
- Department of General Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Zhen-Zhai Cai
- Department of Gastroenterology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
29
|
You Y, Wen R, Pathak R, Li A, Li W, St Clair D, Hauer-Jensen M, Zhou D, Liang Y. Latexin sensitizes leukemogenic cells to gamma-irradiation-induced cell-cycle arrest and cell death through Rps3 pathway. Cell Death Dis 2014; 5:e1493. [PMID: 25341047 PMCID: PMC4237263 DOI: 10.1038/cddis.2014.443] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/04/2014] [Accepted: 09/08/2014] [Indexed: 11/09/2022]
Abstract
Leukemia is a leading cause of cancer death. Recently, the latexin (Lxn) gene was identified as a potential tumor suppressor in several types of solid tumors and lymphoma, and Lxn expression was found to be absent or downregulated in leukemic cells. Whether Lxn functions as a tumor suppressor in leukemia and what molecular and cellular mechanisms are involved are unknown. In this study, the myeloid leukemogenic FDC-P1 cell line was used as a model system and Lxn was ectopically expressed in these cells. Using the protein pull-down assay and mass spectrometry, ribosomal protein subunit 3 (Rps3) was identified as a novel Lxn binding protein. Ectopic expression of Lxn inhibited FDC-P1 growth in vitro. More surprisingly, Lxn enhanced gamma irradiation-induced DNA damages and induced cell-cycle arrest and massive necrosis, leading to depletion of FDC-P1 cells. Mechanistically, Lxn inhibited the nuclear translocation of Rps3 upon radiation, resulting in abnormal mitotic spindle formation and chromosome instability. Rps3 knockdown increased the radiation sensitivity of FDC-P1, confirming that the mechanism of action of Lxn is mediated by Rps3 pathway. Moreover, Lxn enhanced the cytotoxicity of chemotherapeutic agent, VP-16, on FDC-P1 cells. Our study suggests that Lxn itself not only suppresses leukemic cell growth but also potentiates the cytotoxic effect of radio- and chemotherapy on cancer cells. Lxn could be a novel molecular target that improves the efficacy of anti-cancer therapy.
Collapse
Affiliation(s)
- Y You
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - R Wen
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - R Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - A Li
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - W Li
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - D St Clair
- Gratuate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | - M Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - D Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Y Liang
- 1] Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA [2] Gratuate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
30
|
Melanoma epigenetics: novel mechanisms, markers, and medicines. J Transl Med 2014; 94:822-38. [PMID: 24978641 PMCID: PMC4479581 DOI: 10.1038/labinvest.2014.87] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/30/2014] [Accepted: 05/08/2014] [Indexed: 02/07/2023] Open
Abstract
The incidence and mortality rates of cutaneous melanoma continue to increase worldwide, despite the deployment of targeted therapies. Recently, there has been rapid growth and development in our understanding of epigenetic mechanisms and their role in cancer pathobiology. Epigenetics--defined as the processes resulting in heritable changes in gene expression beyond those caused by alterations in the DNA sequence--likely contain the information that encodes for such phenotypic variation between individuals with identical genotypes. By altering the structure of chromatin through covalent modification of DNA bases or histone proteins, or by regulating mRNA translation through non-coding RNAs, the epigenome ultimately determines which genes are expressed and which are kept silent. While our understanding of epigenetic mechanisms is growing at a rapid pace, the field of melanoma epigenomics still remains in its infancy. In this Pathology in Focus, we will briefly review the basics of epigenetics to contextualize and critically examine the existing literature using melanoma as a cancer paradigm. Our understanding of how dysregulated DNA methylation and DNA demethylation/hydroxymethylation, histone modification, and non-coding RNAs affect cancer pathogenesis and melanoma virulence, in particular, provides us with an ever-expanding repertoire of potential diagnostic biomarkers, therapeutic targets, and novel pathogenic mechanisms. The evidence reviewed herein indicates the critical role of epigenetic mechanisms in melanoma pathobiology and provides evidence for future targets in the development of next-generation biomarkers and therapeutics.
Collapse
|
31
|
Latexin exhibits tumor suppressor potential in hepatocellular carcinoma. Oncol Rep 2014; 31:1364-72. [PMID: 24399246 DOI: 10.3892/or.2014.2966] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/16/2013] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver and latexin is downregulated in several types of human cancer. However, latexin expression in HCC remains unknown. mRNA expression of latexin in HCC samples and HCC-derived cell lines was detected by semi‑quantitative PCR and real-time PCR, while protein expression was assessed by immunohistochemistry. The role of latexin in the regulation of the proliferation of HCC-derived cells was investigated both in vitro and in vivo. Flow cytometry was used to differentiate cell cycle distribution in SK-hep-1 and YY-8103. In a total of 60 paired HCC specimens, compared with adjacent non-cancer tissues, latexin mRNA was downregulated in 42 specimens. Immunohistochemical analysis showed a significant reduction in latexin expression in HCC compared to control tissues. Overexpression of latexin inhibited SK-hep-1 and HepG2 cellular colony formation and tumor growth. Conversely, YY‑8103 and Focus cells transfected with shRNA enhanced colony formation and tumor growth. Latexin overexpression promoted cell cycle arrest in the G0/G1 phase in SK-hep-1 and silencing of latexin promoted the cell cycle transition from G0/G1 phase to S phase in YY-8103. The cyclin-dependent kinase inhibitors (CDKIs) (p21Cip1, p27Kip1, p15INK4B), cyclin D1 and cyclin E were shown to be differentially expressed in latexin-overexpressed cells and latexin-silenced cells. These results indicated that latexin may be an effective target for gene therapy.
Collapse
|
32
|
Fukada SI, Ma Y, Ohtani T, Watanabe Y, Murakami S, Yamaguchi M. Isolation, characterization, and molecular regulation of muscle stem cells. Front Physiol 2013; 4:317. [PMID: 24273513 PMCID: PMC3824104 DOI: 10.3389/fphys.2013.00317] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/14/2013] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle has great regenerative capacity which is dependent on muscle stem cells, also known as satellite cells. A loss of satellite cells and/or their function impairs skeletal muscle regeneration and leads to a loss of skeletal muscle power; therefore, the molecular mechanisms for maintaining satellite cells in a quiescent and undifferentiated state are of great interest in skeletal muscle biology. Many studies have demonstrated proteins expressed by satellite cells, including Pax7, M-cadherin, Cxcr4, syndecan3/4, and c-met. To further characterize satellite cells, we established a method to directly isolate satellite cells using a monoclonal antibody, SM/C-2.6. Using SM/C-2.6 and microarrays, we measured the genes expressed in quiescent satellite cells and demonstrated that Hesr3 may complement Hesr1 in generating quiescent satellite cells. Although Hesr1- or Hesr3-single knockout mice show a normal skeletal muscle phenotype, including satellite cells, Hesr1/Hesr3-double knockout mice show a gradual decrease in the number of satellite cells and increase in regenerative defects dependent on satellite cell numbers. We also observed that a mouse's genetic background affects the regenerative capacity of its skeletal muscle and have established a line of DBA/2-background mdx mice that has a much more severe phenotype than the frequently used C57BL/10-mdx mice. The phenotype of DBA/2-mdx mice also seems to depend on the function of satellite cells. In this review, we summarize the methodology of direct isolation, characterization, and molecular regulation of satellite cells based on our results. The relationship between the regenerative capacity of satellite cells and progression of muscular disorders is also summarized. In the last part, we discuss application of the accumulating scientific information on satellite cells to treatment of patients with muscular disorders.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University Osaka, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Zhang H, Ren Y, Xu H, Pang D, Duan C, Liu C. The expression of stem cell protein Piwil2 and piR-932 in breast cancer. Surg Oncol 2013; 22:217-23. [PMID: 23992744 DOI: 10.1016/j.suronc.2013.07.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND To investigate the expression status of PIWIL2 and piR-932 in breast cancer stem cells and the role they could play in tumor cell growth and metastasis through Latexin. METHODS CD44(+)/CD24(-) tumor cells (CSC) from clinical specimens were sorted using flow cytometry. PIWIL2 expression status was detected in CSC cells by microarray analysis and 1086 breast cancer specimens by Western blot and immunohistochemistry staining. piR-932 expression was also detected in CSC cells by piRNA microarray assay. The relationship between the PIWIL2 protein and clinico-pathological parameters and prognosis was subsequently determined. RESULTS CSC cells are more likely to generate new tumors in mice and cell microspheres that are deficient in NOD/SCID compared to the control group. PIWIL2 protein was expressed higher in CSC cells compared to the control cells. In total, 334 (30.76%) of the 1086 breast cases showed high PIWIL2 expression. PIWIL2 was observed to be related to age, tumor size, histological type, tumor stage, and lymph node metastasis (all P < 0.05). Furthermore, we have found that one of the Piwi-interacting RNAs (piRNAs) called piR-932 expressed significantly higher in the breast cancer cells that were induced to EMT, and it could form immune complexes through immunoprecipitation with PIWIL2; in PIWIL2+ breast cancer stem cells, Latexin expression significantly reduced because of its promoter region CpG island methylation. CONCLUSIONS These results suggest that the combination of piR-932 and PIWIL2 may be a positive regulator in the process of breast cancer stem cells through promoting the methylation of Latexin, and they both could be the potential targets for blocking the metastasis of breast cancer.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Breast Surgery, Second Hospital of Dalian Medical University, Dalian 116023, China.
| | | | | | | | | | | |
Collapse
|
34
|
Kocerha J, Liu Y, Willoughby D, Chidamparam K, Benito J, Nelson K, Xu Y, Chi T, Engelhardt H, Moran S, Yang SH, Li SH, Li XJ, Larkin K, Neumann A, Banta H, Yang JJ, Chan AWS. Longitudinal transcriptomic dysregulation in the peripheral blood of transgenic Huntington's disease monkeys. BMC Neurosci 2013; 14:88. [PMID: 23957861 PMCID: PMC3751855 DOI: 10.1186/1471-2202-14-88] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 08/14/2013] [Indexed: 12/30/2022] Open
Abstract
Background Huntington’s Disease (HD) is a progressive neurodegenerative disorder caused by an expansion in the polyglutamine (polyQ) region of the Huntingtin (HTT) gene. The clinical features of HD are characterized by cognitive, psychological, and motor deficits. Molecular instability, a core component in neurological disease progression, can be comprehensively evaluated through longitudinal transcriptomic profiling. Development of animal models amenable to longitudinal examination enables distinct disease-associated mechanisms to be identified. Results Here we report the first longitudinal study of transgenic monkeys with genomic integration of various lengths of the human HTT gene and a range of polyQ repeats. With this unique group of transgenic HD nonhuman primates (HD monkeys), we profiled over 47,000 transcripts from peripheral blood collected over a 2 year timespan from HD monkeys and age-matched wild-type control monkeys. Conclusions Messenger RNAs with expression patterns which diverged with disease progression in the HD monkeys considerably facilitated our search for transcripts with diagnostic or therapeutic potential in the blood of human HD patients, opening up a new avenue for clinical investigation.
Collapse
|
35
|
Wang Y, Chen X, Tsai S, Thomas A, Shizuru JA, Cao TM. Fine mapping of the Bmgr5 quantitative trait locus for allogeneic bone marrow engraftment in mice. Immunogenetics 2013; 65:585-96. [PMID: 23666360 PMCID: PMC3713196 DOI: 10.1007/s00251-013-0709-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/26/2013] [Indexed: 01/04/2023]
Abstract
To identify novel mechanisms regulating allogeneic hematopoietic cell engraftment, we used forward genetics and previously described identification, in mice, of a bone marrow (BM) engraftment quantitative trait locus (QTL), termed Bmgr5. This QTL confers dominant and large allele effects for engraftment susceptibility. It was localized to chromosome 16 by quantitative genetic techniques in a segregating backcross bred from susceptible BALB.K and resistant B10.BR mice. We now report verification of the Bmgr5 QTL using reciprocal chromosome 16 consomic strains. The BM engraftment phenotype in these consomic mice shows that Bmgr5 susceptibility alleles are not only sufficient but also indispensable for conferring permissiveness for allogeneic BM engraftment. Using panels of congenic mice, we resolved the Bmgr5 QTL into two separate subloci, termed Bmgr5a (Chr16:14.6-15.8 Mb) and Bmgr5b (Chr16:15.8-17.6 Mb), each conferring permissiveness for the engraftment phenotype and both fine mapped to an interval amenable to positional cloning. Candidate Bmgr5 genes were then prioritized using whole exome DNA sequencing and microarray gene expression data. Further studies are warranted to elucidate the genetic interaction between the Bmgr5a and Bmgr5b QTL and identify causative genes and underlying gene variants. This may lead to new approaches for overcoming the problem of graft rejection in clinical hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Blood and Marrow Transplantation Program, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Xinjian Chen
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Schickwann Tsai
- Blood and Marrow Transplantation Program, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Alun Thomas
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Judith A. Shizuru
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Thai M. Cao
- Blood and Marrow Transplantation Program, Department of Medicine, University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
36
|
Dzieciatkowska M, Silliman CC, Moore EE, Kelher MR, Banerjee A, Land KJ, Ellison M, West FB, Ambruso DR, Hansen KC. Proteomic analysis of the supernatant of red blood cell units: the effects of storage and leucoreduction. Vox Sang 2013; 105:210-8. [PMID: 23663258 DOI: 10.1111/vox.12042] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/15/2013] [Accepted: 03/21/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND Red blood cell (RBC) transfusion is a life-saving intervention for critically ill patients; however, it has been linked to increased morbidity and mortality. We hypothesize that a number of important proteins accumulate during routine storage of RBCs, which may explain some of the adverse effects seen in transfused patients. STUDY DESIGN Five RBC units were drawn and divided (half prestorage leucoreduced (LR-RBC) and half left as an unmodified control (RBC). The supernatant was separated on days 1 and 42 of storage and proteomic analyses completed with in-gel tryptic digestion and nano-liquid chromatography tandem mass spectrometry. RESULTS In RBC supernatants, 401 proteins were identified: 203 increased with storage, 114 decreased, and 84 were unchanged. In LR-RBC supernatant, 231 proteins were identified: 84 increased with storage, 30 decreased, and 117 were unchanged. Prestorage leucoreduction removed many platelet- and leucocyte-derived structural proteins; however, a number of intracellular proteins accumulated including peroxiredoxins (Prdx) 6 and latexin. The increases were confirmed by immunoblotting, including the T-phosphorylation of Prdx-6, indicating that it may be functioning as an active phospholipase. Active matrix metalloproteinase-9 also increased with a coinciding decrease in the metalloproteinase inhibitor 1 and cystatin C. CONCLUSION We conclude that a number of proteins increase with RBC storage, which is partially ameliorated with leucoreduction, and transfusion of stored RBCs may introduce mediators that result in adverse events in the transfused host.
Collapse
Affiliation(s)
- M Dzieciatkowska
- Departments of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Oldridge EE, Walker HF, Stower MJ, Simms MS, Mann VM, Collins AT, Pellacani D, Maitland NJ. Retinoic acid represses invasion and stem cell phenotype by induction of the metastasis suppressors RARRES1 and LXN. Oncogenesis 2013; 2:e45. [PMID: 23588494 PMCID: PMC3641360 DOI: 10.1038/oncsis.2013.6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mouse haematopoietic stem cell (SC) regulator Latexin (LXN) is the only known homologue of the retinoic acid receptor responder 1 (RARRES1) gene. Both genes lie adjacent on chromosome 3 and differ mostly by the presence of a transmembrane domain in RARRES1. Despite their homology, it is not known whether they possess similar regulatory mechanisms, cellular localization and function. Here, we identified RARRES1 and LXN as highly significantly downregulated genes in human prostate SCs, whose expression was induced by the pro-differentiation agent all-trans retinoic acid (atRA). AtRA induced expression in the most differentiated cells compared with the SC fraction, suggesting that this subpopulation was less responsive to atRA. Small interfering RNA suppression of RARRES1 and LXN enhanced the SC properties of primary prostate cultures, as shown by a significant increase in their colony-forming ability. Expression of both RARRES1 and LXN was co-ordinately repressed by DNA methylation in prostate cancer cell lines and inhibition of RARRES1 and LXN increased the invasive capacity of primary prostate cultures, which also fully rescued an inhibitory effect induced by atRA. Moreover, we showed that RARRES1 and LXN reside within different sub-cellular compartments, providing evidence that RARRES1 is not a plasma membrane protein as previously supposed but is located primarily in the endoplasmic reticulum; whereas LXN was detected in the nucleus of prostate epithelial cells. Thus, LXN and RARRES1 are potential tumour suppressor genes, which are co-ordinately regulated, SC-silenced genes functioning to suppress invasion and colony-forming ability of prostate cancer cells; yet the proteins reside within different sub-cellular compartments.
Collapse
Affiliation(s)
- E E Oldridge
- YCR Cancer Research Unit, Department of Biology, University of York, York, UK
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Muthusamy V, Premi S, Soper C, Platt J, Bosenberg M. The hematopoietic stem cell regulatory gene latexin has tumor-suppressive properties in malignant melanoma. J Invest Dermatol 2013; 133:1827-33. [PMID: 23364479 PMCID: PMC3683103 DOI: 10.1038/jid.2013.48] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite recent advancements in therapy, melanoma still remains a highly lethal skin cancer. A better understanding of the genetic and epigenetic changes responsible for melanoma formation and progression could result in development of more effective treatments. Advanced melanomas are known to exhibit widespread promoter region CpG island methylation leading to inactivation of key tumor suppressor genes. Meta-analyses of relevant microarray data sets revealed the hematopoietic stem cell regulator gene Latexin (LXN) to be commonly down regulated in approximately 50% of melanomas. The CpG island in the promoter region of LXN was almost universally hypermethylated in melanoma cell lines and tumors and treatment of the cell lines with the demethylating drug, 5-Aza-2-deoxycytidine, resulted in increased LXN expression. In this paper, we demonstrate that exogenous expression of LXN in melanoma cell lines results in a significant inhibition of tumor cell proliferation. In addition, we show that the increased expression of LXN in these lines correlates with reduction in expression levels of stem cell transcription factors OCT4, NANOG, SOX2, KLF4 and MYCN indicating that LXN may exert its tumor suppressive function by altering the stem cell like properties of melanoma cells.
Collapse
Affiliation(s)
- Viswanathan Muthusamy
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | |
Collapse
|
39
|
Peng C, Chen Y, Shan Y, Zhang H, Guo Z, Li D, Li S. LSK derived LSK- cells have a high apoptotic rate related to survival regulation of hematopoietic and leukemic stem cells. PLoS One 2012; 7:e38614. [PMID: 22675576 PMCID: PMC3366951 DOI: 10.1371/journal.pone.0038614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 05/07/2012] [Indexed: 11/19/2022] Open
Abstract
A balanced pool of hematopoietic stem cells (HSCs) in bone marrow is tightly regulated, and this regulation is disturbed in hematopoietic malignancies such as chronic myeloid leukemia (CML). The underlying mechanisms are largely unknown. Here we show that the Lin−Sca-1+c-Kit- (LSK−) cell population derived from HSC-containing Lin−Sca-1+c-Kit+ (LSK) cells has significantly higher numbers of apoptotic cells. Depletion of LSK cells by radiation or the cytotoxic chemical 5-fluorouracil results in an expansion of the LSK− population. In contrast, the LSK− population is reduced in CML mice, and depletion of leukemia stem cells (LSCs; BCR-ABL-expressing HSCs) by deleting Alox5 or by inhibiting heat shock protein 90 causes an increase in this LSK− population. The transition of LSK to LSK− cells is controlled by the Icsbp gene and its downstream gene Lyn, and regulation of this cellular transition is critical for the survival of normal LSK cells and LSCs. These results indicate a potential function of the LSK− cells in the regulation of LSK cells and LSCs.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Ly/metabolism
- Apoptosis/drug effects
- Apoptosis/radiation effects
- Arachidonate 5-Lipoxygenase/metabolism
- Benzamides
- Cell Lineage/drug effects
- Cell Lineage/radiation effects
- Cell Survival/drug effects
- Cell Survival/radiation effects
- Fluorouracil/pharmacology
- Fusion Proteins, bcr-abl/metabolism
- Gamma Rays
- HSP90 Heat-Shock Proteins/metabolism
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Hematopoietic Stem Cells/radiation effects
- Imatinib Mesylate
- Interferon Regulatory Factors/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Membrane Proteins/metabolism
- Mice
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/radiation effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Proto-Oncogene Proteins c-kit/metabolism
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Receptors, Cell Surface/metabolism
- Signal Transduction/drug effects
- Signal Transduction/radiation effects
- Signaling Lymphocytic Activation Molecule Family Member 1
- Time Factors
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Cong Peng
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yaoyu Chen
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yi Shan
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Haojian Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Zhiru Guo
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Dongguang Li
- School of Computer and Security Science, Edith Cowan University, Mount Lawley, Western Australia, Australia
| | - Shaoguang Li
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Mitsunaga K, Kikuchi J, Wada T, Furukawa Y. Latexin regulates the abundance of multiple cellular proteins in hematopoietic stem cells. J Cell Physiol 2012; 227:1138-47. [PMID: 21567403 DOI: 10.1002/jcp.22834] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Latexin is the only known carboxypeptidase A inhibitor in mammals and shares structural similarity with cystatin C, suggesting that latexin regulates the abundance of as yet unidentified target proteins. A forward genetic approach revealed that latexin is involved in homeostasis of hematopoietic stem cells (HSCs) in mice; however, little is known about the mechanisms by which latexin negatively affects the numbers of HSCs. In this study, we found that latexin is preferentially expressed in hematopoietic stem/progenitor cells, and is co-localized with the molecules responsible for the interaction of HSCs with a bone marrow niche, such as N-cadherin, Tie2, and Roundabout 4. Latexin-knockout young female mice showed an increase in the numbers of KSL (c-Kit(+)/Sca-1(+)/linegae marker-negative) cells, which may be attributable to enhanced self-renewal because latexin-deficient KSL cells formed more colonies than their wild-type counterparts in methylcellulose culture. Proteomic analysis of Sca-1(+) bone marrow cells demonstrated that latexin ablation reduced the abundance of multiple cellular proteins, including N-cadherin, Tie2, and Roundabout 4. Finally, we found that latexin expression was lost or greatly reduced in approximately 50% of human leukemia/lymphoma cell lines. These results imply that latexin inhibits the self-renewal of HSCs by facilitating the lodgment of HSCs within a bone marrow niche to maintain HSC homeostasis.
Collapse
Affiliation(s)
- Kanae Mitsunaga
- Division of Stem Cell Regulation, Center for Molecular Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | | | | | | |
Collapse
|
41
|
Avagyan S, Aguilo F, Kamezaki K, Snoeck HW. Quantitative trait mapping reveals a regulatory axis involving peroxisome proliferator-activated receptors, PRDM16, transforming growth factor-β2 and FLT3 in hematopoiesis. Blood 2011; 118:6078-86. [PMID: 21967974 PMCID: PMC3234666 DOI: 10.1182/blood-2011-07-365080] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/23/2011] [Indexed: 01/21/2023] Open
Abstract
Hematopoiesis is the process whereby BM HSCs renew to maintain their number or to differentiate into committed progenitors to generate all blood cells. One approach to gain mechanistic insight into this complex process is the investigation of quantitative genetic variation in hematopoietic function among inbred mouse strains. We previously showed that TGF-β2 is a genetically determined positive regulator of hematopoiesis. In the presence of unknown nonprotein serum factors TGF-β2, but not TGF-β1 or -β3, enhances progenitor proliferation in vitro, an effect that is subject to mouse strain-dependent variation mapping to a locus on chr.4, Tb2r1. TGF-β2-deficient mice show hematopoietic defects, demonstrating the physiologic role of this cytokine. Here, we show that TGF-β2 specifically and predominantly cell autonomously enhances signaling by FLT3 in vitro and in vivo. A coding polymorphism in Prdm16 (PR-domain-containing 16) underlies Tb2r1 and differentially regulates transcriptional activity of peroxisome proliferator-activated receptor-γ (PPARγ), identifying lipid PPAR ligands as the serum factors required for regulation of FLT3 signaling by TGF-β2. We furthermore show that PPARγ agonists play a FLT3-dependent role in stress responses of progenitor cells. These observations identify a novel regulatory axis that includes PPARs, Prdm16, and TGF-β2 in hematopoiesis.
Collapse
Affiliation(s)
- Serine Avagyan
- Children's Hospital of New York-Presbyterian, Columbia University Medical Center, New York, NY, USA
| | | | | | | |
Collapse
|
42
|
G protein-coupled receptor kinase 5 mediates Tazarotene-induced gene 1-induced growth suppression of human colon cancer cells. BMC Cancer 2011; 11:175. [PMID: 21575264 PMCID: PMC3112162 DOI: 10.1186/1471-2407-11-175] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/17/2011] [Indexed: 01/08/2023] Open
Abstract
Background Tazarotene-induced gene 1 (TIG1) is a retinoid-inducible type II tumour suppressor gene. The B isoform of TIG1 (TIG1B) inhibits growth and invasion of cancer cells. Expression of TIG1B is frequently downregulated in various cancer tissues; however, the expression and activities of the TIG1A isoform are yet to be reported. Therefore, this study investigated the effects of the TIG1A and TIG1B isoforms on cell growth and gene expression profiles using colon cancer cells. Methods TIG1A and TIG1B stable clones derived from HCT116 and SW620 colon cancer cells were established using the GeneSwitch system; TIG1 isoform expression was induced by mifepristone treatment. Cell growth was assessed using the WST-1 cell proliferation and colony formation assays. RNA interference was used to examine the TIG1 mediating changes in cell growth. Gene expression profiles were determined using microarray and validated using real-time polymerase chain reaction, and Western blot analyses. Results Both TIG1 isoforms were expressed at high levels in normal prostate and colon tissues and were downregulated in colon cancer cell lines. Both TIG1 isoforms significantly inhibited the growth of transiently transfected HCT116 cells and stably expressing TIG1A and TIG1B HCT116 and SW620 cells. Expression of 129 and 55 genes was altered upon induction of TIG1A and TIG1B expression, respectively, in stably expressing HCT116 cells. Of the genes analysed, 23 and 6 genes were upregulated and downregulated, respectively, in both TIG1A and TIG1B expressing cells. Upregulation of the G-protein-coupled receptor kinase 5 (GRK5) was confirmed using real-time polymerase chain reaction and Western blot analyses in both TIG1 stable cell lines. Silencing of TIG1A or GRK5 expression significantly decreased TIG1A-mediated cell growth suppression. Conclusions Expression of both TIG1 isoforms was observed in normal prostate and colon tissues and was downregulated in colon cancer cell lines. Both TIG1 isoforms suppressed cell growth and stimulated GRK5 expression in HCT116 and SW620 cells. Knockdown of GRK5 expression alleviated TIG1A-induced growth suppression of HCT116 cells, suggesting that GRK5 mediates cell growth suppression by TIG1A. Thus, TIG1 may participate in the downregulation of G-protein coupled signaling by upregulating GRK5 expression.
Collapse
|
43
|
Li Y, Basang Z, Ding H, Lu Z, Ning T, Wei H, Cai H, Ke Y. Latexin expression is downregulated in human gastric carcinomas and exhibits tumor suppressor potential. BMC Cancer 2011; 11:121. [PMID: 21466706 PMCID: PMC3080345 DOI: 10.1186/1471-2407-11-121] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 04/06/2011] [Indexed: 01/10/2023] Open
Abstract
Background Latexin, also known as endogenous carboxypeptidase inhibitor (CPI), has been found to inhibit mouse stem cell populations and lymphoma cell proliferation, demonstrating its potential role as a tumor suppressor. Our previous study also suggested a correlation between latexin expression and malignant transformation of immortalized human gastric epithelial cells. Here, we examined latexin expression in human gastric carcinomas and investigated the effect of differential latexin expression on proliferation of gastric cancer cells in vitro and in vivo. Methods Monoclonal antibody against human latexin was prepared and immunohistochemical analysis was performed to detect latexin expression in 41 paired gastric carcinomas and adjacent normal control tissues. Human gastric cancer cells MGC803 (latexin negative) stably transfected with LXN gene and BGC823 cells (latexin positive) stably transfected with antisense LXN gene were established for anchorage-dependent colony formation assay and tumorigenesis assay in nude mice. Differentially expressed genes in response to exogeneous latexin expression were screened using microarray analysis and identified by RT-PCR. Bisulfite sequencing was performed to analyze the correlation of the methylation status of LXN promoter with latexin expression in cell lines. Results Immunohistochemical analysis showed significantly reduced latexin expression in gastric carcinomas (6/41, 14.6%) compared to control tissues (31/41, 75.6%) (P < 0.05). Overexpression of LXN gene in MGC803 cells inhibited colony formation and tumor growth in nude mice. Conversely, BGC823 cells transfected with antisense LXN gene exhibited enhanced tumor growth and colony formation. Additionally, several tumor related genes, including Maspin, WFDC1, SLPI, S100P, and PDGFRB, were shown to be differentially expressed in MGC803 cells in response to latexin expression. Differential expression of Maspin and S100P was also identified in BGC823 cells while latexin expression was downregulated. Further bisulfite sequencing of the LXN gene promoter indicated CpG hypermethylation was correlated with silencing of latexin expression in human cells. Conclusions Latexin expression was reduced in human gastric cancers compared with their normal control tissues. The cellular and molecular evidences demonstrated the inhibitory effect of latexin in human gastric cancer cell growth and tumorigenicity. These results strongly suggest the possible involvement of latexin expression in tumor suppression.
Collapse
Affiliation(s)
- Yong Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Genetics, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Hai Dian District, Beijing 100142, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Sahab ZJ, Hall MD, Me Sung Y, Dakshanamurthy S, Ji Y, Kumar D, Byers SW. Tumor suppressor RARRES1 interacts with cytoplasmic carboxypeptidase AGBL2 to regulate the α-tubulin tyrosination cycle. Cancer Res 2011; 71:1219-28. [PMID: 21303978 DOI: 10.1158/0008-5472.can-10-2294] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Even though it is among the most commonly methylated loci in multiple cancers, the retinoic acid-induced tumor suppressor retinoic acid receptor responder 1 (RARRES1) has no known function. We now show that RARRES1 is lost in many cancer cells, particularly those with a mesenchymal phenotype, and is a transmembrane carboxypeptidase inhibitor that interacts with ATP/GTP binding protein-like 2 (AGBL2), a cytoplasmic carboxypeptidase. Knockdown of AGBL2 results in a failure of the cell to detyrosinate the C-terminal EEY region of α-tubulin and indicates that it is a candidate for the long sought-after tubulin tyrosine carboxypeptidase important in the regulation of microtubule dynamics. In contrast, knockdown of RARRES1 increases the level of detyrosinated α-tubulin consistent with a role as the cognate inhibitor of AGBL2. We conclude that RARRES1, its interacting partners AGBL2, Eg5/KIF11, another EEY-bearing protein (EB1), and the microtubule tyrosination cycle are important in tumorigenesis and identify a novel area for therapeutic intervention.
Collapse
Affiliation(s)
- Ziad J Sahab
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Kawauchi S, Terasaki H, Katano M, Murase J, Masuda Y, Tamura T, Shimadzu M. Quality control and monitoring for the isolation process of mesenchymal stem cells and their differentiation into osteoblasts. Genet Test Mol Biomarkers 2010; 14:269-82. [PMID: 20187761 DOI: 10.1089/gtmb.2009.0139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We developed a method of quality control and monitoring for the isolation of mesenchymal stem cells (MSCs) from bone marrow and their differentiation into osteoblasts. After dividing the cell culture process into five groups based on cell types such as MSCs and osteoblasts, we used microarray analysis to select genes with expression profiles characteristic of each group and quantitative polymerase chain reaction for confirming the expression profiles of these genes. Comparing multiple gene expression profiles per cell from quantitative polymerase chain reaction permitted us to distinguish (1) different groups of cell culture including MSCs and osteoblasts; (2) MSCs that had differentiated cells other than osteoblasts such as chondroblasts, adipocytes, or skin-derived fibroblasts; and (3) desirable MSCs from undesirable MSCs occurring under different culture conditions. These findings suggest that it is possible to standardize MSCs and osteoblasts on the basis of multiple gene expression profiles and to check the quality of these cells. We believe that our methods can be applied to cells cultured for transplants.
Collapse
|
46
|
Kumar R, Avagyan S, Snoeck HW. A quantitative trait locus on chr.4 regulates thymic involution. J Gerontol A Biol Sci Med Sci 2010; 65:620-5. [PMID: 20371546 DOI: 10.1093/gerona/glq041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The mechanisms underlying age-associated thymic involution are unknown. In mice, thymic involution shows mouse strain-dependent genetic variation. Identification of the underlying genes would provide mechanistic insight into this elusive process. We previously showed that responsiveness of hematopoietic stem and progenitor cells (HSPCs) to transforming growth factor-beta 2, a positive regulator of HSPC proliferation, is regulated by a quantitative trait locus (QTL) on chr. 4, Tb2r1. Interestingly, Tgfb2(+/-) mice have delayed thymic involution. Therefore, we tested the hypothesis that a QTL on chr. 4 might regulate thymic involution. Aged, but not young, B6.D2-chr.4 congenic mice, where the telomeric region of chr. 4 was introgressed from DBA/2 to C57BL/6 mice, had larger thymi, and better maintenance of early thymic precursors than C57BL/6 control mice. These observations unequivocally demonstrate that the telomeric region of chr. 4 contains a QTL, Ti1 (thymic involution 1) that regulates thymic involution, and suggest the possibility that Ti1 may be identical to Tb2r1.
Collapse
Affiliation(s)
- Ritu Kumar
- Department of Gene and Cell Medicine, Mount Sinai of School of Medicine, Gustave L. Levy Place, PO Box 1496, New York, NY 10029, USA
| | | | | |
Collapse
|
47
|
Fukada SI, Morikawa D, Yamamoto Y, Yoshida T, Sumie N, Yamaguchi M, Ito T, Miyagoe-Suzuki Y, Takeda S, Tsujikawa K, Yamamoto H. Genetic background affects properties of satellite cells and mdx phenotypes. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2414-24. [PMID: 20304955 DOI: 10.2353/ajpath.2010.090887] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common lethal genetic disorder of children. The mdx (C57BL/10 background, C57BL/10-mdx) mouse is a widely used model of DMD, but the histopathological hallmarks of DMD, such as the smaller number of myofibers, accumulation of fat and fibrosis, and insufficient regeneration of myofibers, are not observed in adult C57BL/10-mdx except for in the diaphragm. In this study, we showed that DBA/2 mice exhibited decreased muscle weight, as well as lower myofiber numbers after repeated degeneration-regeneration cycles. Furthermore, the self-renewal efficiency of satellite cells of DBA/2 is lower than that of C57BL/6. Therefore, we produced a DBA/2-mdx strain by crossing DBA/2 and C57BL/10-mdx. The hind limb muscles of DBA/2-mdx mice exhibited lower muscle weight, fewer myofibers, and increased fat and fibrosis, in comparison with C57BL/10-mdx. Moreover, remarkable muscle weakness was observed in DBA/2-mdx. These results indicate that the DBA/2-mdx mouse is a more suitable model for DMD studies, and the efficient satellite cell self-renewal ability of C57BL/10-mdx might explain the difference in pathologies between humans and mice.
Collapse
Affiliation(s)
- So-ichiro Fukada
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Milner LC, Buck KJ. Identifying quantitative trait loci (QTLs) and genes (QTGs) for alcohol-related phenotypes in mice. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 91:173-204. [PMID: 20813243 DOI: 10.1016/s0074-7742(10)91006-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alcoholism is a complex clinical disorder with genetic and environmental contributions. Although no animal model duplicates alcoholism, models for specific factors, such as the withdrawal syndrome, are useful to identify potential genetic determinants of liability in humans. Murine models have been invaluable to identify quantitative trait loci (QTLs) that influence a variety of alcohol responses. However, the QTL regions are typically large, at least initially, and contain numerous genes, making identification of the causal quantitative trait gene(s) (QTGs) challenging. Here, we present QTG identification strategies currently used in the field of alcohol genetics and discuss relevance to alcoholic human populations.
Collapse
Affiliation(s)
- Lauren C Milner
- Department of Behavioral Neuroscience, VA Medical Center and Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|
49
|
Overall RW, Kempermann G, Peirce J, Lu L, Goldowitz D, Gage FH, Goodwin S, Smit AB, Airey DC, Rosen GD, Schalkwyk LC, Sutter TR, Nowakowski RS, Whatley S, Williams RW. Genetics of the hippocampal transcriptome in mouse: a systematic survey and online neurogenomics resource. Front Neurosci 2009; 3:55. [PMID: 20582282 PMCID: PMC2858614 DOI: 10.3389/neuro.15.003.2009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 10/26/2009] [Indexed: 11/13/2022] Open
Abstract
Differences in gene expression in the CNS influence behavior and disease susceptibility. To systematically explore the role of normal variation in expression on hippocampal structure and function, we generated an online microarray database for a diverse panel of strains of mice, including most common inbred strains and numerous recombinant inbred lines (www.genenetwork.org). Using this resource, coexpression networks for families of genes can be generated rapidly to test causal models related to function. The data set is optimized for quantitative trait locus (QTL) mapping and was used to identify over 5500 QTLs that modulate mRNA levels. We describe a wide variety of analyses and novel synthetic approaches that take advantage of this resource, and demonstrate how both the data and associated tools can be applied to the study of gene regulation in the hippocampus and relations to structure and function.
Collapse
Affiliation(s)
- Rupert W Overall
- Genomics of Regeneration, DFG Research Center for Regenerative Therapies Dresden, Technische Universität Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Van Zant G, Liang Y. Natural genetic diversity as a means to uncover stem cell regulatory pathways. Ann N Y Acad Sci 2009; 1176:170-7. [PMID: 19796245 DOI: 10.1111/j.1749-6632.2009.04567.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Natural genetic diversity is a largely untapped reservoir for use in the discovery of stem cell regulatory pathways. Here we explore the means by which phenotypic diversity in mice can lead to the discovery of novel genes affecting stem cell regulation. We use as an example the discovery that latexin is a regulator of the natural size of the hematopoietic stem cell population in mice. The fact that it is a negative regulator of stem cell numbers, and thus served as a brake on stem cell expansion, led us to consider the possibility that it acts as a tumor suppressor. Experimental evidence supporting this hypothesis is reviewed.
Collapse
Affiliation(s)
- Gary Van Zant
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536-0093, USA.
| | | |
Collapse
|