1
|
Shenoy US, Adiga D, Alhedyan F, Kabekkodu SP, Radhakrishnan R. HOXA9 transcription factor is a double-edged sword: from development to cancer progression. Cancer Metastasis Rev 2024; 43:709-728. [PMID: 38062297 PMCID: PMC11156722 DOI: 10.1007/s10555-023-10159-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/30/2023] [Indexed: 04/02/2024]
Abstract
The HOXA9 transcription factor serves as a molecular orchestrator in cancer stemness, epithelial-mesenchymal transition (EMT), metastasis, and generation of the tumor microenvironment in hematological and solid malignancies. However, the multiple modes of regulation, multifaceted functions, and context-dependent interactions responsible for the dual role of HOXA9 as an oncogene or tumor suppressor in cancer remain obscure. Hence, unravelling its molecular complexities, binding partners, and interacting signaling molecules enables us to comprehend HOXA9-mediated transcriptional programs and molecular crosstalk. However, it is imperative to understand its central role in fundamental biological processes such as embryogenesis, foetus implantation, hematopoiesis, endothelial cell proliferation, and tissue homeostasis before designing targeted therapies. Indeed, it presents an enormous challenge for clinicians to selectively target its oncogenic functions or restore tumor-suppressive role without altering normal cellular functions. In addition to its implications in cancer, the present review also focuses on the clinical applications of HOXA9 in recurrence and drug resistance, which may provide a broader understanding beyond oncology, open new avenues for clinicians for accurate diagnoses, and develop personalized treatment strategies. Furthermore, we have also discussed the existing therapeutic options and accompanying challenges in HOXA9-targeted therapies in different cancer types.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Faisal Alhedyan
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
2
|
Liao Z, Tang S, Nozawa K, Shimada K, Ikawa M, Monsivais D, Matzuk M. Affinity-tagged SMAD1 and SMAD5 mouse lines reveal transcriptional reprogramming mechanisms during early pregnancy. eLife 2024; 12:RP91434. [PMID: 38536963 PMCID: PMC10972565 DOI: 10.7554/elife.91434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Endometrial decidualization, a prerequisite for successful pregnancies, relies on transcriptional reprogramming driven by progesterone receptor (PR) and bone morphogenetic protein (BMP)-SMAD1/SMAD5 signaling pathways. Despite their critical roles in early pregnancy, how these pathways intersect in reprogramming the endometrium into a receptive state remains unclear. To define how SMAD1 and/or SMAD5 integrate BMP signaling in the uterus during early pregnancy, we generated two novel transgenic mouse lines with affinity tags inserted into the endogenous SMAD1 and SMAD5 loci (Smad1HA/HA and Smad5PA/PA). By profiling the genome-wide distribution of SMAD1, SMAD5, and PR in the mouse uterus, we demonstrated the unique and shared roles of SMAD1 and SMAD5 during the window of implantation. We also showed the presence of a conserved SMAD1, SMAD5, and PR genomic binding signature in the uterus during early pregnancy. To functionally characterize the translational aspects of our findings, we demonstrated that SMAD1/5 knockdown in human endometrial stromal cells suppressed expressions of canonical decidual markers (IGFBP1, PRL, FOXO1) and PR-responsive genes (RORB, KLF15). Here, our studies provide novel tools to study BMP signaling pathways and highlight the fundamental roles of SMAD1/5 in mediating both BMP signaling pathways and the transcriptional response to progesterone (P4) during early pregnancy.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Graduate Program of Genetics and Genomics, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Martin Matzuk
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Graduate Program of Genetics and Genomics, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
3
|
Liao Z, Tang S, Nozawa K, Shimada K, Ikawa M, Monsivais D, Matzuk MM. Affinity-tagged SMAD1 and SMAD5 mouse lines reveal transcriptional reprogramming mechanisms during early pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.25.559321. [PMID: 38106095 PMCID: PMC10723262 DOI: 10.1101/2023.09.25.559321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Endometrial decidualization, a prerequisite for successful pregnancies, relies on transcriptional reprogramming driven by progesterone receptor (PR) and bone morphogenetic protein (BMP)-SMAD1/SMAD5 signaling pathways. Despite their critical roles in early pregnancy, how these pathways intersect in reprogramming the endometrium into a receptive state remains unclear. To define how SMAD1 and/or SMAD5 integrate BMP signaling in the uterus during early pregnancy, we generated two novel transgenic mouse lines with affinity tags inserted into the endogenous SMAD1 and SMAD5 loci (Smad1HA/HA and Smad5PA/PA). By profiling the genome-wide distribution of SMAD1, SMAD5, and PR in the mouse uterus, we demonstrated the unique and shared roles of SMAD1 and SMAD5 during the window of implantation. We also showed the presence of a conserved SMAD1, SMAD5, and PR genomic binding signature in the uterus during early pregnancy. To functionally characterize the translational aspects of our findings, we demonstrated that SMAD1/5 knockdown in human endometrial stromal cells suppressed expressions of canonical decidual markers (IGFBP1, PRL, FOXO1) and PR-responsive genes (RORB, KLF15). Here, our studies provide novel tools to study BMP signaling pathways and highlight the fundamental roles of SMAD1/5 in mediating both BMP signaling pathways and the transcriptional response to progesterone (P4) during early pregnancy.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Martin M. Matzuk
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
4
|
Agrawal-Singh S, Bagri J, Giotopoulos G, Azazi DMA, Horton SJ, Lopez CK, Anand S, Bach AS, Stedham F, Antrobus R, Houghton JW, Vassiliou GS, Sasca D, Yun H, Whetton AD, Huntly BJP. HOXA9 forms a repressive complex with nuclear matrix-associated protein SAFB to maintain acute myeloid leukemia. Blood 2023; 141:1737-1754. [PMID: 36577137 PMCID: PMC10113176 DOI: 10.1182/blood.2022016528] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/07/2022] [Accepted: 11/28/2022] [Indexed: 12/29/2022] Open
Abstract
HOXA9 is commonly upregulated in acute myeloid leukemia (AML), in which it confers a poor prognosis. Characterizing the protein interactome of endogenous HOXA9 in human AML, we identified a chromatin complex of HOXA9 with the nuclear matrix attachment protein SAFB. SAFB perturbation phenocopied HOXA9 knockout to decrease AML proliferation, increase differentiation and apoptosis in vitro, and prolong survival in vivo. Integrated genomic, transcriptomic, and proteomic analyses further demonstrated that the HOXA9-SAFB (H9SB)-chromatin complex associates with nucleosome remodeling and histone deacetylase (NuRD) and HP1γ to repress the expression of factors associated with differentiation and apoptosis, including NOTCH1, CEBPδ, S100A8, and CDKN1A. Chemical or genetic perturbation of NuRD and HP1γ-associated catalytic activity also triggered differentiation, apoptosis, and the induction of these tumor-suppressive genes. Importantly, this mechanism is operative in other HOXA9-dependent AML genotypes. This mechanistic insight demonstrates the active HOXA9-dependent differentiation block as a potent mechanism of disease maintenance in AML that may be amenable to therapeutic intervention by targeting the H9SB interface and/or NuRD and HP1γ activity.
Collapse
Affiliation(s)
- Shuchi Agrawal-Singh
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Jaana Bagri
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - George Giotopoulos
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Dhoyazan M A Azazi
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah J Horton
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Cecile K Lopez
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Shubha Anand
- Cancer Molecular Diagnostics Laboratory, Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Anne-Sophie Bach
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Frances Stedham
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jack W Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - George S Vassiliou
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel Sasca
- Department of Hematology, Oncology and Pneumology, University Medical Center Mainz, Mainz, Germany
| | - Haiyang Yun
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Anthony D Whetton
- School of Veterinary Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| | - Brian J P Huntly
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Qian J, LeSavage BL, Hubka KM, Ma C, Natarajan S, Eggold JT, Xiao Y, Fuh KC, Krishnan V, Enejder A, Heilshorn SC, Dorigo O, Rankin EB. Cancer-associated mesothelial cells promote ovarian cancer chemoresistance through paracrine osteopontin signaling. J Clin Invest 2021; 131:e146186. [PMID: 34396988 DOI: 10.1172/jci146186] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/25/2021] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer is the leading cause of gynecological malignancy-related deaths, due to its widespread intraperitoneal metastases and acquired chemoresistance. Mesothelial cells are an important cellular component of the ovarian cancer microenvironment that promote metastasis. However, their role in chemoresistance is unclear. Here, we investigated whether cancer-associated mesothelial cells promote ovarian cancer chemoresistance and stemness in vitro and in vivo. We found that osteopontin is a key secreted factor that drives mesothelial-mediated ovarian cancer chemoresistance and stemness. Osteopontin is a secreted glycoprotein that is clinically associated with poor prognosis and chemoresistance in ovarian cancer. Mechanistically, ovarian cancer cells induced osteopontin expression and secretion by mesothelial cells through TGF-β signaling. Osteopontin facilitated ovarian cancer cell chemoresistance via the activation of the CD44 receptor, PI3K/AKT signaling, and ABC drug efflux transporter activity. Importantly, therapeutic inhibition of osteopontin markedly improved the efficacy of cisplatin in both human and mouse ovarian tumor xenografts. Collectively, our results highlight mesothelial cells as a key driver of ovarian cancer chemoresistance and suggest that therapeutic targeting of osteopontin may be an effective strategy for enhancing platinum sensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Jin Qian
- Department of Radiation Oncology
| | | | - Kelsea M Hubka
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Chenkai Ma
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, New South Wales, Australia
| | | | | | | | - Katherine C Fuh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University, St. Louis, Missouri, USA
| | - Venkatesh Krishnan
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Oliver Dorigo
- Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Erinn B Rankin
- Department of Radiation Oncology.,Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| |
Collapse
|
6
|
Amilca-Seba K, Sabbah M, Larsen AK, Denis JA. Osteopontin as a Regulator of Colorectal Cancer Progression and Its Clinical Applications. Cancers (Basel) 2021; 13:cancers13153793. [PMID: 34359694 PMCID: PMC8345080 DOI: 10.3390/cancers13153793] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022] Open
Abstract
Simple Summary The mortality of colorectal cancer is principally related to metastatic disease at the time of diagnosis or to the growth of initially undetectable micro-metastasis. Current therapeutic strategies are efficient in patients with locally advanced cancer, but are rarely able to cure patients with metastatic disease. Therapeutic failure is mainly associated with drug resistance and an aggressive phenotype. The identification of new biomarkers for micro-metastasis and tumor progression remains an unmet clinical need that should allow for improved patient stratification for optimal treatment and may lead to the identification of novel therapeutic targets. Osteopontin (OPN), a multifunctional protein, has emerged as a potentially valuable biomarker in several cancer types. This review principally describes the molecular mechanisms of OPN that are associated with colorectal cancer (CRC) progression and metastasis, as well as the use of OPN as a clinical biomarker. This review identifies a role for OPN as a biomarker ready for extended clinical application and discusses its use as a therapeutic target. Abstract A high expression of the phosphoprotein osteopontin (OPN) has been associated with cancer progression in several tumor types, including breast cancer, hepatocarcinoma, ovarian cancer, and colorectal cancer (CRC). Interestingly, OPN is overexpressed in CRC and is associated with a poor prognosis linked to invasion and metastasis. Here, we review the regulation and functions of OPN with an emphasis on CRC. We examine how epigenetic and genetic regulators interact with the key signaling pathways involved in this disease. Then, we describe the role of OPN in cancer progression, including proliferation, survival, migration, invasion, and angiogenesis. Furthermore, we outline the interest of using OPN as a clinical biomarker, and discuss if and how osteopontin can be implemented as a routine assay in clinical laboratories for monitoring CRC patients. Finally, we discuss the use of OPN an attractive, but challenging, therapeutic target.
Collapse
Affiliation(s)
- Katyana Amilca-Seba
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
| | - Michèle Sabbah
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Annette K. Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Jérôme A. Denis
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Department of Endocrinology and Oncology Biochemistry, Pitié-Salpetrière Hospital, 75013 Paris, France
- Correspondence: ; Tel.: +33-(0)1-42-16-20-39
| |
Collapse
|
7
|
Grand Moursel L, van der Graaf LM, Bulk M, van Roon‐Mom WM, van der Weerd L. Osteopontin and phospho-SMAD2/3 are associated with calcification of vessels in D-CAA, an hereditary cerebral amyloid angiopathy. Brain Pathol 2019; 29:793-802. [PMID: 30868685 PMCID: PMC6850614 DOI: 10.1111/bpa.12721] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/06/2019] [Indexed: 01/06/2023] Open
Abstract
In severe forms of cerebral amyloid angiopathy (CAA) pathology, vascular calcification has been observed in the cerebral cortex, both in vivo on MRI and CT, and post-mortem using histopathology. However, the pathomechanisms leading to calcification of CAA-laden arteries are unknown. Therefore, we investigated the correlation between calcification of cortical arterioles and several potential modulators of vascular calcification using immunohistochemistry in a unique collection of brain material of patients with a hereditary form of CAA, namely hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D or D-CAA). We show a topographical association of osteopontin (OPN) and TGFβ signaling factor phospho-SMAD2/3 (pSMAD2/3) in calcified CAA vessel walls. OPN and pSMAD2/3 gradually accumulate in vessels prior to calcification. Moreover, we found that the vascular accumulation of Collagen 1 (Col1), OPN and pSMAD2/3 immunomarkers correlated with the CAA severity. This was independently of the vessel size, including capillaries in the most severe cases. We propose that calcification of CAA vessels in the observed HCHWA-D cases may be induced by extracellular OPN trapped in the fibrotic Col1 vessel wall, independently of the presence of vascular amyloid.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Linda M. van der Graaf
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Marjolein Bulk
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | | | - Louise van der Weerd
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
8
|
Lambert M, Alioui M, Jambon S, Depauw S, Van Seuningen I, David-Cordonnier MH. Direct and Indirect Targeting of HOXA9 Transcription Factor in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11060837. [PMID: 31213012 PMCID: PMC6627208 DOI: 10.3390/cancers11060837] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 01/14/2023] Open
Abstract
HOXA9 (Homeobox A9) is a homeotic transcription factor known for more than two decades to be associated with leukemia. The expression of HOXA9 homeoprotein is associated with anterior-posterior patterning during embryonic development, and its expression is then abolished in most adult cells, with the exception of hematopoietic progenitor cells. The oncogenic function of HOXA9 was first assessed in human acute myeloid leukemia (AML), particularly in the mixed-phenotype associated lineage leukemia (MPAL) subtype. HOXA9 expression in AML is associated with aggressiveness and a poor prognosis. Since then, HOXA9 has been involved in other hematopoietic malignancies and an increasing number of solid tumors. Despite this, HOXA9 was for a long time not targeted to treat cancer, mainly since, as a transcription factor, it belongs to a class of protein long considered to be an "undruggable" target; however, things have now evolved. The aim of the present review is to focus on the different aspects of HOXA9 targeting that could be achieved through multiple ways: (1) indirectly, through the inhibition of its expression, a strategy acting principally at the epigenetic level; or (2) directly, through the inhibition of its transcription factor function by acting at either the protein/protein interaction or the protein/DNA interaction interfaces.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
- Institut pour la Recherche sur le Cancer de Lille, F-59045 Lille, France.
| | - Meryem Alioui
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
- Institut pour la Recherche sur le Cancer de Lille, F-59045 Lille, France.
| | - Samy Jambon
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
- Institut pour la Recherche sur le Cancer de Lille, F-59045 Lille, France.
| | - Sabine Depauw
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
- Institut pour la Recherche sur le Cancer de Lille, F-59045 Lille, France.
| | - Isabelle Van Seuningen
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
| | - Marie-Hélène David-Cordonnier
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre Aubert Neurosciences and Cancer, F-59000 Lille, France.
- Institut pour la Recherche sur le Cancer de Lille, F-59045 Lille, France.
| |
Collapse
|
9
|
Khaliullin TO, Kisin ER, Murray AR, Yanamala N, Shurin MR, Gutkin DW, Fatkhutdinova LM, Kagan VE, Shvedova AA. Mediation of the single-walled carbon nanotubes induced pulmonary fibrogenic response by osteopontin and TGF-β1. Exp Lung Res 2018; 43:311-326. [PMID: 29140132 DOI: 10.1080/01902148.2017.1377783] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF THE STUDY A number of in vivo studies have shown that pulmonary exposure to carbon nanotubes (CNTs) may lead to an acute local inflammatory response, pulmonary fibrosis, and granulomatous lesions. Among the factors that play direct roles in initiation and progression of fibrotic processes are epithelial-mesenchymal transition and myofibroblasts recruitment/differentiation, both mediated by transforming growth factor-β1 (TGF-β1). Yet, other contributors to TGF-β1 associated signaling, such as osteopontin (OPN) has not been fully investigated. MATERIALS AND METHODS OPN-knockout female mice (OPN-KO) along with their wild-type (WT) counterparts were exposed to single-walled carbon nanotubes (SWCNT) (40 µg/mouse) via pharyngeal aspiration and fibrotic response was assessed 1, 7, and 28 days post-exposure. Simultaneously, RAW 264.7 and MLE-15 cells were treated with SWCNT (24 hours, 6 µg/cm2 to 48 µg/cm2) or bleomycin (0.1 µg/ml) in the presence of OPN-blocking antibody or isotype control, and TGF-β1 was measured in supernatants. RESULTS AND CONCLUSIONS Diminished lactate dehydrogenase activity at all time points, along with less pronounced neutrophil influx 24 h post-exposure, were measured in broncho-alveolar lavage (BAL) of OPN-KO mice compared to WT. Pro-inflammatory cytokine release (IL-6, TNF-α, MCP-1) was reduced. A significant two-fold increase of TGF-β1 was found in BAL of WT mice at 7 days, while TGF-β1 levels in OPN-KO animals remained unaltered. Histological examination revealed marked decrease in granuloma formation and less collagen deposition in the lungs of OPN-KO mice compared to WT. RAW 264.7 but not MLE-15 cells exposed to SWCNT and bleomycin had significantly less TGF-β1 released in the presence of OPN-blocking antibody. We believe that OPN is important in initiating the cellular mechanisms that produce an overall pathological response to SWCNT and it may act upstream of TGF-β1. Further investigation to understand the mechanistic details of such interactions is critical to predict outcomes of pulmonary exposure to CNT.
Collapse
Affiliation(s)
- Timur O Khaliullin
- a Department of Physiology & Pharmacology , West Virginia University , Morgantown , WV.,b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV
| | - Elena R Kisin
- b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV
| | | | | | - Michael R Shurin
- c Department Pathology , University of Pittsburgh , Pittsburgh , PA
| | - Dmitriy W Gutkin
- c Department Pathology , University of Pittsburgh , Pittsburgh , PA
| | - Liliya M Fatkhutdinova
- d Department of Hygiene and Occupational Medicine , Kazan State Medical University , Kazan , Russia
| | - Valerian E Kagan
- e Department of Pathology , University of Pittsburgh , Pittsburgh , PA
| | - Anna A Shvedova
- a Department of Physiology & Pharmacology , West Virginia University , Morgantown , WV.,b Exposure Assessment Branch , NIOSH/CDC , Morgantown , WV
| |
Collapse
|
10
|
Osteopontin-integrin interaction as a novel molecular target for antibody-mediated immunotherapy in adult T-cell leukemia. Retrovirology 2015; 12:99. [PMID: 26597716 PMCID: PMC4657376 DOI: 10.1186/s12977-015-0225-x] [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: 06/12/2015] [Accepted: 11/12/2015] [Indexed: 12/18/2022] Open
Abstract
Background Adult T-cell leukemia (ATL) is a CD4+ T-cell neoplasm with a poor prognosis. A previous study has shown that there is a strong correlation between the secreted matricellular protein osteopontin (OPN) level and disease severity in ATL patients. Here, we investigated the role of OPN in ATL pathogenesis and the possible application of anti-OPN monoclonal antibody (mAb) for ATL immunotherapy in NOD/Shi-scid,IL-2Rgnull (NOG) mice. Results Subcutaneous inoculation of ATL cell lines into NOG mice increased the plasma level of OPN, which significantly correlated with metastasis of the inoculated cells and survival time. Administration of an SVVYGLR motif-recognizing anti-OPN mAb resulted in inhibition not only of tumor growth but also of tumor invasion and metastasis. The number of fibroblast activating protein-positive fibroblasts was also reduced by this mAb. We then co-inoculated mouse embryonic fibroblasts (MEFs) isolated from wild-type (WT) or OPN knockout mice together with ATL-derived TL-OmI cells into the NOG mice. The mice co-inoculated with WT MEFs displayed a significant decrease in survival relative to those injected with TL-OmI cells alone and the absence of OPN in MEFs markedly improved the survival rate of TL-OmI-inoculated mice. In addition, tumor volume and metastasis were also reduced in the absence of OPN. Conclusion We showed that the xenograft NOG mice model can be a useful system for assessment of the physiological role of OPN in ATL pathogenesis. Using this xenograft model, we found that fibroblast-derived OPN was involved in tumor growth and metastasis, and that this tumor growth and metastasis was significantly suppressed by administration of the anti-OPN mAbs. Our findings will lead to a novel mAb-mediated immunotherapeutic strategy targeting against the interaction of OPN with integrins on the tumor of ATL patients. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0225-x) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Down-regulation of osteopontin mediates a novel mechanism underlying the cytostatic activity of TGF-β. Cell Oncol (Dordr) 2015; 39:119-28. [PMID: 26584547 DOI: 10.1007/s13402-015-0257-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Loss of a cytostatic response to TGF-β has been implicated in multiple hyper-proliferative disorders, including cancer. Although several key genes involved in the cytostatic activity of TGF-β have in the past been identified, its exact mode of action is yet to be elucidated. A comprehensive understanding of the mechanisms underlying the cytostatic activity of TGF-β may open up new avenues for the development of therapeutic strategies. METHODS Quantitative real-time RT-PCR was used to assess osteopontin (OPN) gene expression in human hepatoma-derived Huh-7 and lung adenocarcinoma-derived A549 cells. Reporter assays using an OPN promoter-luciferase construct and its mutated counterparts were performed to assess its transcriptional activity. Binding of Smad4 to the OPN gene promoter was investigated using chromatin immunoprecipitation (CHIP). The putative role of Smad4 in OPN gene expression down-regulation was also assessed using a shRNA-mediated knockdown strategy. The anti-proliferative effect of TGF-β on different cancer-derived cell lines was determined using the cell proliferation reagent WST-1. RESULTS We found that the OPN expression levels dose-dependently decreased in TGF-β-treated Huh-7 and A549 cells. Our reporter assays indicated that this TGF-β-induced repression occurred at the transcriptional level, and could largely be abrogated by disruption of an element (TIE2) similar to the TGF-β inhibitory element found in other TGF-β-repressed genes. Our CHIP assay revealed that the Smad protein complex specifically binds to the OPN gene promoter, and that the TGF-β-mediated inhibition of OPN was lost upon shRNA-mediated knockdown of Smad4. Moreover, we found that the deregulation of OPN gene expression by TGF-β occurred concomitantly with loss of the TGF-β anti-proliferative response, whereas a neutralizing anti-OPN antibody partially restored this response. CONCLUSIONS Our results indicate that the OPN gene is a direct target of Smad-mediated TGF-β signaling, implying that OPN expression inhibition serves as a novel mechanism underlying the cytostatic activity of TGF-β.
Collapse
|
12
|
Role of HOXA9 in leukemia: dysregulation, cofactors and essential targets. Oncogene 2015; 35:1090-8. [PMID: 26028034 DOI: 10.1038/onc.2015.174] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/24/2015] [Accepted: 04/14/2015] [Indexed: 02/08/2023]
Abstract
HOXA9 is a homeodomain-containing transcription factor that has an important role in hematopoietic stem cell expansion and is commonly deregulated in acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia lead to overexpression of HOXA9, which is a strong predictor of poor prognosis. In many cases, HOXA9 has been shown to be necessary for maintaining leukemic transformation; however, the molecular mechanisms through which it promotes leukemogenesis remain elusive. Recent work has established that HOXA9 regulates downstream gene expression through binding at promoter distal enhancers along with a subset of cell-specific cofactor and collaborator proteins. Increasing efforts are being made to identify both the critical cofactors and target genes required for maintaining transformation in HOXA9-overexpressing leukemias. With continued advances in understanding HOXA9-mediated transformation, there is a wealth of opportunity for developing novel therapeutics that would be applicable for greater than 50% of AML with overexpression of HOXA9.
Collapse
|
13
|
Song H, Deng B, Zou C, Huai W, Zhao R, Zhao W. GSK3β negatively regulates LPS-induced osteopontin expression via inhibiting its transcription. Scand J Immunol 2015; 81:186-91. [PMID: 25565601 DOI: 10.1111/sji.12268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 12/03/2014] [Indexed: 01/23/2023]
Abstract
Osteopontin (OPN) is expressed by a variety of immune cells and is critical for both innate and adaptive immune responses. The expression status of OPN might be tightly regulated to maintain immune homeostasis. However, the mechanisms by which OPN is negatively regulated in LPS-stimulated macrophages remain largely unknown. In this study, we showed that glycogen synthase kinase 3β (GSK3β) inhibitors - SB216763, LiCl and azakenpaullone - enhanced LPS-induced OPN expression in mouse peritoneal macrophages. GSK3β knock-down had the similar effects. Furthermore, we found that GSK3β inhibitors and GSK3β knock-down both increased the activity of OPN promoter in LPS-stimulated macrophages. GSK3β inhibitor-mediated enhancement of LPS-induced OPN promoter activity was abrogated in GSK3β siRNA-treated macrophages. Therefore, we identified GSK3β as a negative regulator of OPN expression and suggest GSK3β as a potential therapeutic target for the intervention of diseases with uncontrolled OPN production.
Collapse
Affiliation(s)
- H Song
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong, China
| | | | | | | | | | | |
Collapse
|
14
|
Li Y, Chao F, Huang B, Liu D, Kim J, Huang S. HOXC8 promotes breast tumorigenesis by transcriptionally facilitating cadherin-11 expression. Oncotarget 2015; 5:2596-607. [PMID: 24810778 PMCID: PMC4058030 DOI: 10.18632/oncotarget.1841] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cell-cell adhesion molecule cadherin-11(CDH11) is preferentially expressed in basal-like breast cancer cells and facilitates breast cancer cell migration by promoting small GTPase Rac activity. However, how the expression of CDH11 is regulated in breast cancer cells is not understood. Here, we show that CDH11 is transcriptionally controlled by homeobox C8 (HOXC8) in human breast cancer cells. HOXC8 serves as a CDH11-specific transcription factor and binds to the site of nucleotides −196 to −191 in the CDH11 promoter. Depletion of HOXC8 leads to the decrease in anchorage-independent cell growth, cell migration/invasion and spontaneous metastasis of breast cancer cells; however, suppressed tumorigenic events were fully rescued by ectopic CDH11 expression in HOXC8-knockdown cells. These results indicate that HOXC8 impacts breast tumorigenesis through CDH11. The analysis of publically available human breast tumor microarray gene expression database demonstrates a strong positive linear association between HOXC8 and CDH11 expression (ρ = 0.801, p < 0.001). Survival analysis (Kaplan-Meier method, log-rank test) shows that both high HOXC8 and CDH11 expression correlate with poor recurrence-free survival rate of patients. Together, our study suggests that HOXC8 promotes breast tumorigenesis by maintaining high level of CDH11 expression in breast cancer cells.
Collapse
Affiliation(s)
- Yong Li
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | | | | | | | | | | |
Collapse
|
15
|
Ndisang JF, Tiwari S. Featured article: induction of heme oxygenase with hemin improves pericardial adipocyte morphology and function in obese Zucker rats by enhancing proteins of regeneration. Exp Biol Med (Maywood) 2014; 240:45-57. [PMID: 25053781 DOI: 10.1177/1535370214544268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress and inflammation are implicated in tissue remodeling, hypertrophy, and organ malfunction. Since heme-oxygenase (HO) is a cytoprotective enzyme with effects against oxidative stress and inflammation, we investigated the effects of upregulating HO with hemin on adipocyte hypertrophy, proteins of repair/regeneration including beta-catenin, Oct3/4 and Pax2 as well as pro-fibrotic/remodeling proteins like osteopontin and transforming growth factor-beta (TGF-β) in pericardial adipose tissue from obese Zucker rats (ZRs). Treatment with hemin significantly reduced pericardial adipose tissue inflammation/oxidative stress, suppressed osteopontin and TGF-β, and attenuated pericardial adipocyte hypertrophy in obese ZRs. These were associated with enhanced expression of the stem/progenitor-cell marker cKit; the potentiation of several proteins of regeneration including beta-catenin, Oct3/4, Pax2; and improved pericardial adipocyte morphology. Interestingly, the amelioration of adipocyte hypertrophy in hemin-treated animals was accompanied by improved adipocyte function, evidenced by increased levels of pericardial adipose tissue adiponectin. Furthermore, hemin significantly reduced hypertriglyceridemia and hypercholesteromia in obese ZRs. The protective effects of hemin were accompanied by robust potentiation HO activity and the total antioxidant capacity, whereas the co-administration of hemin with the HO inhibitor, stannous mesoporphyrin abolished the effects of hemin. These data suggest that hemin improves pericardial adipocyte morphology and function by enhancing proteins of repair and regeneration, while concomitantly abating inflammatory/oxidative insults and suppressing extracellular-matrix/profibrotic and remodeling proteins. The reduction of hypertriglyceridemia, hypercholesteromia, pericardial adiposity, and pericardial adipocyte hypertrophy with corresponding improvement of adipocyte morphology/function in hemin-treated animals suggests that HO inducers may be explored for the design of novel remedies against cardiac complications arising from excessive adiposity.
Collapse
Affiliation(s)
- Joseph Fomusi Ndisang
- Department of Physiology, College of Medicine, University of Saskatchewan College of Medicine, Saskatoon, SK, Canada S7N 5E5
| | - Shuchita Tiwari
- Department of Physiology, College of Medicine, University of Saskatchewan College of Medicine, Saskatoon, SK, Canada S7N 5E5
| |
Collapse
|
16
|
Ko SY, Naora H. HOXA9 promotes homotypic and heterotypic cell interactions that facilitate ovarian cancer dissemination via its induction of P-cadherin. Mol Cancer 2014; 13:170. [PMID: 25023983 PMCID: PMC4105245 DOI: 10.1186/1476-4598-13-170] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 07/04/2014] [Indexed: 12/30/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) is a lethal disease that frequently involves the peritoneal cavity. Dissemination of EOC is a multi-step process in which exfoliated tumor cells survive in the peritoneal fluid as multi-cellular aggregates and then form invasive implants on peritoneal surfaces. The mechanisms that control this process are poorly understood. We previously identified that high expression of the developmental patterning gene HOXA9 is associated with poor survival in EOC patients. In this study, we investigated the significance and mechanisms of HOXA9 in controlling aggregation and implantation of floating EOC cells. Methods HOXA9 was inhibited by shRNAs or expressed in EOC cells that were propagated in suspension cultures and in the peritoneal cavity of mice. Cell death was assayed by flow cytometry and ELISA. Cell aggregation, attachment and migration were evaluated by microscopy, transwell chamber assays and histopathologic analysis. DNA-binding of HOXA9 and its effect on expression of the cell adhesion molecule P-cadherin were assayed by chromatin immunoprecipitation, quantitative RT-PCR and Western blot. HOXA9 and P-cadherin expression was evaluated in publicly available datasets of EOC clinical specimens. Results We identified that HOXA9 promotes aggregation and inhibits anoikis in floating EOC cells in vitro and in xenograft models. HOXA9 also stimulated the ability of EOC cells to attach to peritoneal cells and to migrate. HOXA9 bound the promoter of the CDH3 gene that encodes P-cadherin, induced CDH3 expression in EOC cells, and was associated with increased CDH3 expression in clinical specimens of EOC. Inhibiting P-cadherin in EOC cells that expressed HOXA9 abrogated the stimulatory effects of HOXA9 on cell aggregation, implantation and migration. Conversely, these stimulatory effects of HOXA9 were restored when P-cadherin was reconstituted in EOC cells in which HOXA9 was inhibited. Conclusion These findings indicate that HOXA9 contributes to poor outcomes in EOC in part by promoting intraperitoneal dissemination via its induction of P-cadherin.
Collapse
Affiliation(s)
| | - Honami Naora
- Department of Molecular & Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Pan G, Cao J, Yang N, Ding K, Fan C, Xiong WC, Hamrick M, Isales CM, Shi XM. Role of glucocorticoid-induced leucine zipper (GILZ) in bone acquisition. J Biol Chem 2014; 289:19373-82. [PMID: 24860090 DOI: 10.1074/jbc.m113.535237] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Glucocorticoids (GCs) have both anabolic and catabolic effects on bone. However, no GC anabolic effect mediator has been identified to date. Here we show that targeted expression of glucocorticoid-induced leucine zipper (GILZ), a GC anti-inflammatory effect mediator, enhances bone acquisition in mice. Transgenic mice, in which the expression of GILZ is under the control of a 3.6-kb rat type I collagen promoter, exhibited a high bone mass phenotype with significantly increased bone formation rate and osteoblast numbers. The increased osteoblast activity correlates with enhanced osteogenic differentiation and decreased adipogenic differentiation of bone marrow stromal cell cultures in vitro. In line with these changes, the mRNA levels of key osteogenic regulators (Runx2 and Osx) increased, and the level of adipogenic regulator peroxisome proliferator-activated receptor (PPAR) γ2 decreased significantly. We also found that GILZ physically interacts with C/EBPs and disrupts C/EBP-mediated PPARγ gene transcription. In conclusion, our results showed that GILZ is capable of increasing bone acquisition in vivo, and this action is mediated via a mechanism involving the inhibition of PPARγ gene transcription and shifting of bone marrow MSC/progenitor cell lineage commitment in favor of the osteoblast pathway.
Collapse
Affiliation(s)
- Guodong Pan
- From the Departments of Neuroscience and Regenerative Medicine, Wuhan University, Wuhan 430072, China, and
| | - Jay Cao
- the Grand Forks Human Nutrition Research Center, United States Department of Agriculture Agricultural Research Service, Grand Forks, North Dakota 58203
| | - Nianlan Yang
- From the Departments of Neuroscience and Regenerative Medicine
| | - Kehong Ding
- From the Departments of Neuroscience and Regenerative Medicine
| | - Cheng Fan
- From the Departments of Neuroscience and Regenerative Medicine
| | - Wen-Cheng Xiong
- From the Departments of Neuroscience and Regenerative Medicine
| | | | - Carlos M Isales
- From the Departments of Neuroscience and Regenerative Medicine, Orthopaedic Surgery,Georgia Regents University, Augusta, Georgia 30912
| | - Xing-Ming Shi
- From the Departments of Neuroscience and Regenerative Medicine, Orthopaedic Surgery,Georgia Regents University, Augusta, Georgia 30912,
| |
Collapse
|
18
|
Schwarz D, Varum S, Zemke M, Schöler A, Baggiolini A, Draganova K, Koseki H, Schübeler D, Sommer L. Ezh2 is required for neural crest-derived cartilage and bone formation. Development 2014; 141:867-77. [PMID: 24496623 DOI: 10.1242/dev.094342] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The emergence of craniofacial skeletal elements, and of the jaw in particular, was a crucial step in the evolution of higher vertebrates. Most facial bones and cartilage are generated during embryonic development by cranial neural crest cells, while an osteochondrogenic fate is suppressed in more posterior neural crest cells. Key players in this process are Hox genes, which suppress osteochondrogenesis in posterior neural crest derivatives. How this specific pattern of osteochondrogenic competence is achieved remains to be elucidated. Here we demonstrate that Hox gene expression and osteochondrogenesis are controlled by epigenetic mechanisms. Ezh2, which is a component of polycomb repressive complex 2 (PRC2), catalyzes trimethylation of lysine 27 in histone 3 (H3K27me3), thereby functioning as transcriptional repressor of target genes. Conditional inactivation of Ezh2 does not interfere with localization of neural crest cells to their target structures, neural development, cell cycle progression or cell survival. However, loss of Ezh2 results in massive derepression of Hox genes in neural crest cells that are usually devoid of Hox gene expression. Accordingly, craniofacial bone and cartilage formation is fully prevented in Ezh2 conditional knockout mice. Our data indicate that craniofacial skeleton formation in higher vertebrates is crucially dependent on epigenetic regulation that keeps in check inhibitors of an osteochondrogenic differentiation program.
Collapse
Affiliation(s)
- Daniel Schwarz
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, CH-8057 Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Knapik DM, Perera P, Nam J, Blazek AD, Rath B, Leblebicioglu B, Das H, Wu LC, Hewett TE, Agarwal SK, Robling AG, Flanigan DC, Lee BS, Agarwal S. Mechanosignaling in bone health, trauma and inflammation. Antioxid Redox Signal 2014; 20:970-85. [PMID: 23815527 PMCID: PMC3924811 DOI: 10.1089/ars.2013.5467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE Mechanosignaling is vital for maintaining the structural integrity of bone under physiologic conditions. These signals activate and suppress multiple signaling cascades regulating bone formation and resorption. Understanding these pathways is of prime importance to exploit their therapeutic potential in disorders associated with bone loss due to disuse, trauma, or disruption of homeostatic mechanisms. RECENT ADVANCES In the case of cells of the bone, an impressive amount of data has been generated that provides evidence of a complex mechanism by which mechanical signals can maintain or disrupt cellular homeostasis by driving transcriptional regulation of growth factors, matrix proteins and inflammatory mediators in health and inflammation. Mechanical signals act on cells in a magnitude dependent manner to induce bone deposition or resorption. During health, physiological levels of these signals are essential for maintaining bone strength and architecture, whereas during inflammation, similar signals can curb inflammation by suppressing the nuclear factor kappa B (NF-κB) signaling cascade, while upregulating matrix synthesis via mothers against decapentaplegic homolog and/or Wnt signaling cascades. Contrarily, excessive mechanical forces can induce inflammation via activation of the NF-κB signaling cascade. CRITICAL ISSUES Given the osteogenic potential of mechanical signals, it is imperative to exploit their therapeutic efficacy for the treatment of bone disorders. Here we review select signaling pathways and mediators stimulated by mechanical signals to modulate the strength and integrity of the bone. FUTURE DIRECTIONS Understanding the mechanisms of mechanotransduction and its effects on bone lay the groundwork for development of nonpharmacologic mechanostimulatory approaches for osteodegenerative diseases and optimal bone health.
Collapse
Affiliation(s)
- Derrick M Knapik
- 1 Department of Orthopaedic Surgery, The Ohio State University College of Medicine , Columbus, Ohio
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Misexpression of Pknox2 in mouse limb bud mesenchyme perturbs zeugopod development and deltoid crest formation. PLoS One 2013; 8:e64237. [PMID: 23717575 PMCID: PMC3661445 DOI: 10.1371/journal.pone.0064237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 04/10/2013] [Indexed: 01/30/2023] Open
Abstract
The TALE (Three Amino acid Loop Extension) family consisting of Meis, Pbx and Pknox proteins is a group of transcriptional co-factors with atypical homeodomains that play pivotal roles in limb development. Compared to the in-depth investigations of Meis and Pbx protein functions, the role of Pknox2 in limb development remains unclear. Here, we showed that Pknox2 was mainly expressed in the zeugopod domain of the murine limb at E10.5 and E11.5. Misexpression of Pknox2 in the limb bud mesenchyme of transgenic mice led to deformities in the zeugopod and forelimb stylopod deltoid crest, but left the autopod and other stylopod skeletons largely intact. These malformations in zeugopod skeletons were recapitulated in mice overexpressing Pknox2 in osteochondroprogenitor cells. Molecular and cellular analyses indicated that the misexpression of Pknox2 in limb bud mesenchyme perturbed the Hox10-11 gene expression profiles, decreased Col2 expression and Bmp/Smad signaling activity in the limb. These results indicated that Pknox2 misexpression affected mesenchymal condensation and early chondrogenic differentiation in the zeugopod skeletons of transgenic embryos, suggesting Pknox2 as a potential regulator of zeugopod and deltoid crest formation.
Collapse
|
21
|
Bandyopadhyay S, Harris DP, Adams GN, Lause GE, McHugh A, Tillmaand EG, Money A, Willard B, Fox PL, DiCorleto PE. HOXA9 methylation by PRMT5 is essential for endothelial cell expression of leukocyte adhesion molecules. Mol Cell Biol 2012; 32:1202-13. [PMID: 22269951 PMCID: PMC3302442 DOI: 10.1128/mcb.05977-11] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 01/12/2012] [Indexed: 11/20/2022] Open
Abstract
The induction of proinflammatory proteins in stimulated endothelial cells (EC) requires activation of multiple transcription programs. The homeobox transcription factor HOXA9 has an important regulatory role in cytokine induction of the EC-leukocyte adhesion molecules (ELAM) E-selectin and vascular cell adhesion molecule 1 (VCAM-1). However, the mechanism underlying stimulus-dependent activation of HOXA9 is completely unknown. Here, we elucidate the molecular mechanism of HOXA9 activation by tumor necrosis factor alpha (TNF-α) and show an unexpected requirement for arginine methylation by protein arginine methyltransferase 5 (PRMT5). PRMT5 was identified as a TNF-α-dependent binding partner of HOXA9 by mass spectrometry. Small interfering RNA (siRNA)-mediated depletion of PRMT5 abrogated stimulus-dependent HOXA9 methylation with concomitant loss in E-selectin or VCAM-1 induction. Chromatin immunoprecipitation analysis revealed that PRMT5 is recruited to the E-selectin promoter following transient HOXA9 binding to its cognate recognition sequence. PRMT5 induces symmetric dimethylation of Arg140 on HOXA9, an event essential for E-selectin induction. In summary, PRMT5 is a critical coactivator component in a newly defined, HOXA9-containing transcription complex. Moreover, stimulus-dependent methylation of HOXA9 is essential for ELAM expression during the EC inflammatory response.
Collapse
Affiliation(s)
- Smarajit Bandyopadhyay
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Daniel P. Harris
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gregory N. Adams
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Gregory E. Lause
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Anne McHugh
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Emily G. Tillmaand
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Angela Money
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Belinda Willard
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Paul L. Fox
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
| | - Paul E. DiCorleto
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
22
|
Lillvis JH, Erdman R, Schworer CM, Golden A, Derr K, Gatalica Z, Cox LA, Shen J, Vander Heide RS, Lenk GM, Hlavaty L, Li L, Elmore JR, Franklin DP, Gray JL, Garvin RP, Carey DJ, Lancaster WD, Tromp G, Kuivaniemi H. Regional expression of HOXA4 along the aorta and its potential role in human abdominal aortic aneurysms. BMC PHYSIOLOGY 2011; 11:9. [PMID: 21627813 PMCID: PMC3125234 DOI: 10.1186/1472-6793-11-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/31/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND The infrarenal abdominal aorta exhibits increased disease susceptibility relative to other aortic regions. Allograft studies exchanging thoracic and abdominal segments showed that regional susceptibility is maintained regardless of location, suggesting substantial roles for embryological origin, tissue composition and site-specific gene expression. RESULTS We analyzed gene expression with microarrays in baboon aortas, and found that members of the HOX gene family exhibited spatial expression differences. HOXA4 was chosen for further study, since it had decreased expression in the abdominal compared to the thoracic aorta. Western blot analysis from 24 human aortas demonstrated significantly higher HOXA4 protein levels in thoracic compared to abdominal tissues (P < 0.001). Immunohistochemical staining for HOXA4 showed nuclear and perinuclear staining in endothelial and smooth muscle cells in aorta. The HOXA4 transcript levels were significantly decreased in human abdominal aortic aneurysms (AAAs) compared to age-matched non-aneurysmal controls (P < 0.00004). Cultured human aortic endothelial and smooth muscle cells stimulated with INF-γ (an important inflammatory cytokine in AAA pathogenesis) showed decreased levels of HOXA4 protein (P < 0.0007). CONCLUSIONS Our results demonstrated spatial variation in expression of HOXA4 in human aortas that persisted into adulthood and that downregulation of HOXA4 expression was associated with AAAs, an important aortic disease of the ageing population.
Collapse
Affiliation(s)
- John H Lillvis
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Roy S, Thakur AR. Two models of Smad4 and Hoxa9 Complex are Proposed: Structural and Interactional Perspective. J Biomol Struct Dyn 2011; 28:729-42. [DOI: 10.1080/07391102.2011.10508602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature 2011; 470:269-73. [PMID: 21289624 DOI: 10.1038/nature09677] [Citation(s) in RCA: 395] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/16/2010] [Indexed: 12/16/2022]
Abstract
Effective clinical management of prostate cancer (PCA) has been challenged by significant intratumoural heterogeneity on the genomic and pathological levels and limited understanding of the genetic elements governing disease progression. Here, we exploited the experimental merits of the mouse to test the hypothesis that pathways constraining progression might be activated in indolent Pten-null mouse prostate tumours and that inactivation of such progression barriers in mice would engender a metastasis-prone condition. Comparative transcriptomic and canonical pathway analyses, followed by biochemical confirmation, of normal prostate epithelium versus poorly progressive Pten-null prostate cancers revealed robust activation of the TGFβ/BMP-SMAD4 signalling axis. The functional relevance of SMAD4 was further supported by emergence of invasive, metastatic and lethal prostate cancers with 100% penetrance upon genetic deletion of Smad4 in the Pten-null mouse prostate. Pathological and molecular analysis as well as transcriptomic knowledge-based pathway profiling of emerging tumours identified cell proliferation and invasion as two cardinal tumour biological features in the metastatic Smad4/Pten-null PCA model. Follow-on pathological and functional assessment confirmed cyclin D1 and SPP1 as key mediators of these biological processes, which together with PTEN and SMAD4, form a four-gene signature that is prognostic of prostate-specific antigen (PSA) biochemical recurrence and lethal metastasis in human PCA. This model-informed progression analysis, together with genetic, functional and translational studies, establishes SMAD4 as a key regulator of PCA progression in mice and humans.
Collapse
|
25
|
Wheadon H, Ramsey JM, Dobbin E, Dickson GJ, Corrigan PM, Freeburn RW, Thompson A. Differential Hox expression in murine embryonic stem cell models of normal and malignant hematopoiesis. Stem Cells Dev 2011; 20:1465-76. [PMID: 21083428 DOI: 10.1089/scd.2010.0226] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The Hox family are master transcriptional regulators of developmental processes, including hematopoiesis. The Hox regulators, caudal homeobox factors (Cdx1-4), and Meis1, along with several individual Hox proteins, are implicated in stem cell expansion during embryonic development, with gene dosage playing a significant role in the overall function of the integrated Hox network. To investigate the role of this network in normal and aberrant, early hematopoiesis, we employed an in vitro embryonic stem cell differentiation system, which recapitulates mouse developmental hematopoiesis. Expression profiles of Hox, Pbx1, and Meis1 genes were quantified at distinct stages during the hematopoietic differentiation process and compared with the effects of expressing the leukemic oncogene Tel/PDGFRβ. During normal differentiation the Hoxa cluster, Pbx1 and Meis1 predominated, with a marked reduction in the majority of Hox genes (27/39) and Meis1 occurring during hematopoietic commitment. Only the posterior Hoxa cluster genes (a9, a10, a11, and a13) maintained or increased expression at the hematopoietic colony stage. Cdx4, Meis1, and a subset of Hox genes, including a7 and a9, were differentially expressed after short-term oncogenic (Tel/PDGFRβ) induction. Whereas Hoxa4-10, b1, b2, b4, and b9 were upregulated during oncogenic driven myelomonocytic differentiation. Heterodimers between Hoxa7/Hoxa9, Meis1, and Pbx have previously been implicated in regulating target genes involved in hematopoietic stem cell (HSC) expansion and leukemic progression. These results provide direct evidence that transcriptional flux through the Hox network occurs at very early stages during hematopoietic differentiation and validates embryonic stem cell models for gaining insights into the genetic regulation of normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Helen Wheadon
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Smad proteins are intracellular molecules that mediate the canonical signaling cascade of TGFbeta superfamily growth factors. The TGFbeta superfamily comprises two groups of growth factors, BMPs and TGFbetas. Both groups can be further divided into several sub-groups based on sequence homologies and functional similarities. Ligands of the TGFbeta superfamily bind to cell surface receptors to activate Smad proteins in the cytoplasm; then the activated Smad proteins translocate into the nucleus to activate or repress specific target gene transcription. Both groups of growth factors play important roles in skeletal development and regeneration. However, whether these effects reflect signaling through canonical Smad pathways, or other non-canonical signaling pathways in vivo remains a mystery. Moreover, the mechanisms utilized by Smad proteins to initiate nuclear events and their interactions with cytoplasmic proteins are still under intensive investigation. This review will discuss the most recent progress understanding Smad signaling in the context of skeletal development and regeneration.
Collapse
Affiliation(s)
- Buer Song
- Orthopedic Hospital Research Center, Department of Orthopedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, United States
| | | | | |
Collapse
|
27
|
Persistent inflammation and angiogenesis during wound healing in K14-directed Hoxb13 transgenic mice. J Invest Dermatol 2009; 130:856-65. [PMID: 19759546 DOI: 10.1038/jid.2009.305] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Chronic, nonhealing wounds and inadequate tissue repair characterized by excessive fibrosis continue to have a considerable negative effect on health and quality of life. Understanding the molecular events required for adequate healing, including the transcriptional control of wound repair, will be important for the development of future therapies. We previously showed that loss of Hoxb13 from murine skin results in enhanced cutaneous wound healing, suggesting that Hoxb13 has a negative effect on wound repair. To test this, we generated skin-specific Hoxb13 transgenic (TG) mice that overexpress Hoxb13 in the basal layer of the epidermis by the human keratin 14 promoter. Using these mice, we evaluated the effects of Hoxb13 overexpression on cutaneous wound healing. Transgenic wounds were characterized by persistence of the fibrin clot and prolonged inflammation. Notably, neutrophils, which had cleared from wild-type wounds, were still pronounced in TG wounds. Marked epidermal hyperplasia was observed at TG wound edges, and dermal vessels were grossly abnormal compared with wild-type mice. Both vascular endothelial growth factor and tumor necrosis factor-alpha were upregulated in Hoxb13 TG skin. Together, our results identify Hoxb13 as a potential important clinical target in wound healing and other pathologies characterized by abnormal or excessive inflammation, angiogenesis, or epidermal proliferation.
Collapse
|
28
|
Abstract
MicroRNAs (miRNAs) are a class of highly conserved small noncoding RNAs that negatively regulate gene expression by imperfectly base pairing to the 3'-untranslated region of their target mRNAs, leading to mRNA degradation or translational inhibition. The emerging field of miRNA biology has begun to unravel roles for these regulatory molecules in a variety of biological processes. This review concentrates on the roles of miRNAs in skeletogenesis as well as in skeleton-related disease processes. Before describing these data, we present a brief review of the biogenesis and action of miRNAs, the approaches to miRNAs study, and miRNAs as global regulators of development. We finish by emphasizing that the study of the biological functions of miRNAs in skeletogenesis and dysplasia represents an entirely new avenue in the exploration of bone and cartilage biology, and large gaps remain in our knowledge of miRNAs in skeletogenesis in vivo and in our knowledge of the molecular events underlying miRNA-mediated musculoskeletal disorders.
Collapse
Affiliation(s)
- Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, New York 10003, USA.
| |
Collapse
|
29
|
Mann RS, Lelli KM, Joshi R. Hox specificity unique roles for cofactors and collaborators. Curr Top Dev Biol 2009; 88:63-101. [PMID: 19651302 DOI: 10.1016/s0070-2153(09)88003-4] [Citation(s) in RCA: 265] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hox proteins are well known for executing highly specific functions in vivo, but our understanding of the molecular mechanisms underlying gene regulation by these fascinating proteins has lagged behind. The premise of this review is that an understanding of gene regulation-by any transcription factor-requires the dissection of the cis-regulatory elements that they act upon. With this goal in mind, we review the concepts and ideas regarding gene regulation by Hox proteins and apply them to a curated list of directly regulated Hox cis-regulatory elements that have been validated in the literature. Our analysis of the Hox-binding sites within these elements suggests several emerging generalizations. We distinguish between Hox cofactors, proteins that bind DNA cooperatively with Hox proteins and thereby help with DNA-binding site selection, and Hox collaborators, proteins that bind in parallel to Hox-targeted cis-regulatory elements and dictate the sign and strength of gene regulation. Finally, we summarize insights that come from examining five X-ray crystal structures of Hox-cofactor-DNA complexes. Together, these analyses reveal an enormous amount of flexibility into how Hox proteins function to regulate gene expression, perhaps providing an explanation for why these factors have been central players in the evolution of morphological diversity in the animal kingdom.
Collapse
Affiliation(s)
- Richard S Mann
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | | | | |
Collapse
|
30
|
Chung ACK, Huang XR, Zhou L, Heuchel R, Lai KN, Lan HY. Disruption of the Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in mice. Nephrol Dial Transplant 2008; 24:1443-54. [PMID: 19096081 DOI: 10.1093/ndt/gfn699] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The present study tested the hypothesis that disruption of Smad7 function may accelerate renal fibrosis and inflammation. METHODS This was investigated in a unilateral ureteral obstruction (UUO) model induced in wild-type (WT) and Smad7DeltaE1 mice in which functional Smad7 is disrupted by deleting exon I in the Smad7 gene. Renal fibrosis and inflammation after UUO were examined by histology, real-time PCR, western blot analyses and immunohistochemistry. RESULTS Seven days after UUO, severe tubulointerstitial fibrosis developed in WT mice as evidenced by a marked increase in alpha-SMA, collagen I and III extracellular matrix. This was associated with a significant upregulation of renal TGF-beta1 and CTGF and activation of Smad2/3. Interestingly, compared to WT UUO mice, Smad7DeltaE1 mice with UUO exhibited a further increase in TGF-beta/Smad2/3-dependent renal fibrosis. Moreover, compared to WT UUO mice, deletion of the Smad7 gene also sustained NF-kappaB activation and thus enhanced further renal inflammation such as macrophage infiltration and upregulation of TNF-alpha, MCP-1, OPN and ICAM-1. CONCLUSION Smad7 is a critical negative regulator of TGF-beta/Smad2/3 and NF-kappaB signalling and plays a negative regulating role in both renal fibrosis and inflammation after UUO. Results from this study further support the notion that Smad7 may be a therapeutic agent for kidney diseases.
Collapse
Affiliation(s)
- Arthur C K Chung
- Department of Medicine, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Osteopontin is a secreted phosphoprotein that has been implicated as an important mediator of tumor metastasis and has been investigated for use as a biomarker for advanced disease and as a potential therapeutic target in the regulation of cancer metastasis. The OPN DNA sequence is highly conserved and the protein contains several important functional domains including alpha(v)beta integrin and CD44 binding sites. High levels of OPN expression correlate with tumor invasion, progression or metastasis in multiple cancer. Studies demonstrate that osteopontin mediates the molecular mechanisms which determine metastatic spread, such as prevention of apoptosis, extracellular matrix proteolysis and remodeling, cell migration, evasion of host-immune cells and neovascularization. Transcriptional regulation of OPN is complex and involves multiple pathways, including AP-1, Myc, v-Src, Runx/CBF, TGF-B/BMPs/Smad/Hox, and Wnt/ss-catenin/APC/GSK-3ss/Tcf-4. The current state of knowledge of OPN biology suggests that it is an attractive target for therapeutic modulation of metastatic disease.
Collapse
|
32
|
Trivedi CM, Patel RC, Patel CV. Homeobox gene HOXA9 inhibits nuclear factor-kappa B dependent activation of endothelium. Atherosclerosis 2007; 195:e50-60. [PMID: 17586512 DOI: 10.1016/j.atherosclerosis.2007.04.055] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 04/05/2007] [Accepted: 04/30/2007] [Indexed: 10/23/2022]
Abstract
Cytokine-induced expression of adhesion molecules such as ICAM-1, VCAM-1, and E-selectin, on activated endothelial cells (EC) plays an essential role in the development of inflammatory diseases like atherosclerosis. Transcription factor nuclear factor-kappa B (NF-kappaB) is mainly responsible for the induced expression of these adhesion molecules in response to pro-inflammatory cytokines. The mechanisms that maintain EC in a "basal" state and negatively regulate EC activation remain to be characterized. HOXA9 is a homeobox transcription factor expressed in EC and its expression is rapidly down-regulated in response to inflammatory signals. In the present study, we demonstrate that HOXA9 overexpression inhibits the induction of ICAM-1, VCAM-1, and E-selectin in response to pro-inflammatory cytokines. HOXA9 inhibits the adhesion molecule expression by inhibiting NF-kappaB dependent transcriptional activation of these promoters. HOXA9 inhibits EC activation downstream of NF-kappaB nuclear localization by interfering with NF-kappaB DNA binding, but not transactivation capacity. Trichostatin A (TSA) rescues HOXA9 mediated suppression of NF-kappaB activity, but HOXA9 interaction with p300 is not responsible for inhibition of EC activation. Thus, our results suggest involvement of HOXA9 in maintaining the "basal" state of EC and demonstrate that downregulation of HOXA9 is an essential event during EC activation in response to inflammatory signals.
Collapse
Affiliation(s)
- Chinmay M Trivedi
- Department of Cell and Developmental Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC-29209, USA
| | | | | |
Collapse
|
33
|
Bandyopadhyay S, Ashraf MZ, Daher P, Howe PH, DiCorleto PE. HOXA9 participates in the transcriptional activation of E-selectin in endothelial cells. Mol Cell Biol 2007; 27:4207-16. [PMID: 17452460 PMCID: PMC1900059 DOI: 10.1128/mcb.00052-07] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The homeobox gene HOXA9 has recently been shown to be an important regulator of endothelial cell (EC) differentiation and activation in addition to its role in embryonic development and hematopoiesis. In this report, we have determined that the EC-leukocyte adhesion molecule E-selectin is a key target for HOXA9. The depletion of HOXA9 protein in ECs resulted in a significant and specific decrease in tumor necrosis factor alpha (TNF-alpha)-induced E-selectin gene expression. In addition, HOXA9 specifically activated the E-selectin gene promoter in ECs. Progressive deletional analyses together with site-specific mutagenesis of the E-selectin promoter indicated that the Abd-B-like HOX DNA-binding motif, CAATTTTATTAA, located in the proximal region spanning bp -210 to -221 upstream of the transcription start site was crucial for the promoter induction by HOXA9. Both HOXA9 in EC nuclear extract and recombinant HOXA9 protein bound to this sequence in vitro. Moreover, we showed that HOXA9 binds temporally, in a TNF-alpha-dependent manner, to the region containing this Abd-B-like element in vivo. We have thus identified a novel and functionally critical cis-regulatory element for TNF-alpha-mediated transient expression of the E-selectin gene. Further, we provide evidence that HOXA9 acts as an obligate proinflammatory factor by mediating cytokine induction of E-selectin.
Collapse
Affiliation(s)
- Smarajit Bandyopadhyay
- Department of Cell Biology, Lerner Research Institute and Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
34
|
Jalvy S, Renault MA, Lam Shang Leen L, Belloc I, Reynaud A, Gadeau AP, Desgranges C. CREB mediates UTP-directed arterial smooth muscle cell migration and expression of the chemotactic protein osteopontin via its interaction with activator protein-1 sites. Circ Res 2007; 100:1292-9. [PMID: 17413042 DOI: 10.1161/01.res.0000266609.28312.de] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The transcription factor cAMP responsive element-binding protein (CREB) has been found to be involved in arterial smooth muscle cell (SMC) migration. We previously demonstrated that osteopontin (OPN) expression is a key step for UTP-mediated migration of arterial SMCs and that activator protein (AP)-1, nuclear factor kappaB, and upstream stimulatory transcription factors are involved in this OPN expression. The present study aims to determine the role of CREB in UTP-induced migration and OPN expression in cultured SMCs. We found that CREB is activated by UTP via extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase pathways but not by protein kinase A. Both overexpression of a dominant negative CREB and CREB small interfering RNA treatment suppressed UTP-induced OPN expression and SMC migration. Gel-shift and chromatin immunoprecipitation assays revealed that CREB binds 2 AP-1 sites (-1870 and -76) and a cAMP responsive element-like site (-1403) on the OPN promoter. Mutations of these sites showed that only the 2 AP-1 sites were required for UTP-induced OPN expression. Moreover, gel-supershift and sequential chromatin immunoprecipitation assays suggested that CREB was associated with c-Fos on the AP-1 sites of the OPN promoter. These results demonstrate that CREB participates in the induction of UTP-activated OPN expression via its binding to 2 AP-1 sites and is thus involved in UTP-mediated SMC migration.
Collapse
|
35
|
Gnainsky Y, Kushnirsky Z, Bilu G, Hagai Y, Genina O, Volpin H, Bruck R, Spira G, Nagler A, Kawada N, Yoshizato K, Reinhardt DP, Libermann TA, Pines M. Gene expression during chemically induced liver fibrosis: effect of halofuginone on TGF-beta signaling. Cell Tissue Res 2006; 328:153-66. [PMID: 17180598 DOI: 10.1007/s00441-006-0330-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Accepted: 08/14/2006] [Indexed: 01/07/2023]
Abstract
Hepatic fibrosis is associated with the activation of stellate cells (HSCs), the major source of extracellular matrix (ECM) proteins. Transforming growth factor-beta (TGF-beta), signaling via Smad3, is the most profibrogenic cytokine and the major promoter of ECM synthesis. Halofuginone, an inhibitor of liver fibrosis, inhibits TGF-beta-dependent Smad3 phosphorylation in human HSCs in culture. We have used transcriptional profiling to evaluate the effect of halofuginone on gene expression during the progression of thioacetamide (TAA)-induced liver fibrosis in the rat and have focused on genes that are associated with TGF-beta. TAA treatment causes alterations in the expression of 7% of liver genes. Halofuginone treatment prevents the changes in the expression of 41% of these genes and results in the inhibition of HSC activation and collagen synthesis. During the early stages of the disease, halofuginone affects genes involved in alcohol, lipid, protein, and phosphate metabolism and cell adhesion and, at later stages, in the cell cycle (cell development, differentiation, cell proliferation, and apoptosis). The activation of TGF-beta-dependent genes, such as tartrate-resistant acid phosphatase, its putative substrate osteopontin, stellate cell activation-association protein, and fibrillin-1, during chemically induced fibrosis is prevented by halofuginone. This study thus highlights the role of TGF-beta signaling in liver fibrosis and especially its potential for pharmacological intervention. Halofuginone, which has demonstrated efficacy and tolerance in animals and humans, could become an effective and novel therapy for liver fibrosis.
Collapse
Affiliation(s)
- Y Gnainsky
- Institute of Animal Sciences, The Volcani Center, P.O. Box 6, Bet Dagan 50250, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Homeodomain proteins of the Hox family play an important role in regulation of normal hematopoiesis. Substantial evidence also indicates that abnormal Hox protein expression is functionally significant in the pathogenesis of acute myeloid malignancies. The purpose of this review is to outline recent progress in understanding molecular mechanisms involved in Hox regulation of myelopoiesis and myeloid leukemogenesis. RECENT FINDINGS Since Hox proteins function as transcription factors, recent studies have focused on identifying Hox target genes. Various approaches to this problem have been taken, including high throughput screening techniques. In these studies, expression profiles of hematopoietic cells overexpressing various Hox proteins have been analyzed to obtain initial information about potential target genes. Identification of common and unique sets of target genes for various Hox proteins will shed light on function and regulation of the Hox code in developing hematopoietic cells. SUMMARY Recent studies have generated some intriguing information about potential Hox target genes involved in myelopoiesis and leukemogenesis. A number of issues regarding Hox protein function are unresolved, however. These issues include determining whether the effects of various Hox proteins are redundant versus antagonistic, identifying mechanisms which regulate Hox protein function and mechanisms by which Hox proteins modulate target gene transcription in a context-dependent manner.
Collapse
Affiliation(s)
- Elizabeth A Eklund
- The Feinberg School of Medicine, Northwestern University and Jesse Brown VHA Medical Center, 710 N. Fairbanks Court, Chicago, IL 60611, USA.
| |
Collapse
|
37
|
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor (TGF)-beta superfamily of signal molecules that mediate many diverse biological processes ranging from early embryonic tissue patterning to postnatal tissue homeostasis. BMPs trigger cell responses mainly through the canonical signaling pathway where intracellular Smads play central roles in delivering the extracellular signals to the nucleus. While the same Smads are used by BMPs in all types of cells, different transcription factors account in part for the functional diversity of BMPs. These transcription factors are recruited by Smads to regulate the expression of specific subsets of target genes depending on the cell types. Among the transcription factors are Hox proteins. Experimental gain and loss-of-function studies as well as naturally occurring mutations in Hox genes demonstrate their central roles in embryonic skeletal patterning. In addition to the interactions with Smads observed for several Hox proteins, there is also evidence that the expression of a number of Hox genes is regulated by BMPs. It is suggested that Hox proteins play an important role in the BMP pathway.
Collapse
Affiliation(s)
- Xuelin Li
- Department of Pathology, University of Alabama at Birmingham, 1670 University Blvd., VHG003, Birmingham, AL 35294, USA
| | | |
Collapse
|
38
|
Svingen T, Tonissen KF. Hox transcription factors and their elusive mammalian gene targets. Heredity (Edinb) 2006; 97:88-96. [PMID: 16721389 DOI: 10.1038/sj.hdy.6800847] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Hox family of homeodomain transcription factors regulate numerous pathways during developmental and normal cellular processes. All Hox proteins recognise similar sequences in vitro yet display functional diversity in an in vivo environment. This review focuses on the transcriptional and functional specificity elicited by Hox proteins, giving an overview of homeodomain-DNA interactions and the gain of binding specificity through cooperative binding with cofactors. Furthermore, currently identified mammalian Hox target genes are presented, of which the most striking feature is that very few direct Hox targets have been identified. The direct targets participate in an array of cellular functions including organogenesis and cellular differentiation, cell adhesion and migration and cell cycle and apoptotic pathways. A further assessment of identified mammalian promoter targets and the contribution of bases outside the canonical recognition motif is given, highlighting roles they may play in either trans-activation or repression by Hox proteins.
Collapse
Affiliation(s)
- T Svingen
- Cell Biology Group, Eskitis Institute for Cell and Molecular Therapies and School of Biomolecular and Biomedical Science, Griffith University, Nathan, Queensland 4111, Australia
| | | |
Collapse
|
39
|
Pearson JC, Lemons D, McGinnis W. Modulating Hox gene functions during animal body patterning. Nat Rev Genet 2006; 6:893-904. [PMID: 16341070 DOI: 10.1038/nrg1726] [Citation(s) in RCA: 618] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With their power to shape animal morphology, few genes have captured the imagination of biologists as the evolutionarily conserved members of the Hox clusters have done. Recent research has provided new insight into how Hox proteins cause morphological diversity at the organismal and evolutionary levels. Furthermore, an expanding collection of sequences that are directly regulated by Hox proteins provides information on the specificity of target-gene activation, which might allow the successful prediction of novel Hox-response genes. Finally, the recent discovery of microRNA genes within the Hox gene clusters indicates yet another level of control by Hox genes in development and evolution.
Collapse
Affiliation(s)
- Joseph C Pearson
- Section in Cell & Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
40
|
Wang N, Kim HG, Cotta CV, Wan M, Tang Y, Klug CA, Cao X. TGFbeta/BMP inhibits the bone marrow transformation capability of Hoxa9 by repressing its DNA-binding ability. EMBO J 2006; 25:1469-80. [PMID: 16525506 PMCID: PMC1440313 DOI: 10.1038/sj.emboj.7601037] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Accepted: 02/14/2006] [Indexed: 11/09/2022] Open
Abstract
Homeobox (Hox) gene mutations and their altered expressions are frequently linked to human leukemia. Here, we report that transforming growth factor beta (TGFbeta)/bone morphogenetic protein (BMP) inhibits the bone marrow transformation capability of Hoxa9 and Nup98-Hoxa9, the chimeric fusion form of Hoxa9 identified in human acute myeloid leukemia (AML), through Smad4, the common Smad (Co-Smad) in the TGFbeta/BMP signaling pathway. Smad4 interacts directly with the homeodomain of Hoxa9 and blocks the ability of Nup98-Hoxa9 to bind DNA, thereby suppressing its ability to regulate downstream gene transcription. Mapping data revealed that the amino-terminus of Smad4 mediates this interaction and overexpression of the Hoxa9 interaction domain of Smad4 was sufficient to inhibit the enhanced serial replating ability of primary bone marrow cells induced by Nup98-Hoxa9. These studies establish a novel mechanism by which TGFbeta/BMP regulates hematopoiesis and suggest that modification of Hox DNA-binding activity may serve as a novel therapeutic intervention for those leukemias that involve deregulation of Hox.
Collapse
Affiliation(s)
- Ning Wang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hyung-Gyoong Kim
- Department of Microbiology, Division of Developmental and Clinical Immunology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Claudiu V Cotta
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Microbiology, Division of Developmental and Clinical Immunology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mei Wan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yi Tang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christopher A Klug
- Department of Microbiology, Division of Developmental and Clinical Immunology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xu Cao
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
41
|
Bondos SE, Tan XX, Matthews KS. Physical and genetic interactions link hox function with diverse transcription factors and cell signaling proteins. Mol Cell Proteomics 2006; 5:824-34. [PMID: 16455680 DOI: 10.1074/mcp.m500256-mcp200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Positional information provided by Hox homeotic transcription factors is integrated with other transcription factors and cell signaling cascades in specific combinations to dictate context- and gene-specific Hox activity. Protein-protein interactions between these groups have long been hypothesized to modulate Hox functions, yielding a context-specific function. However, difficulties in applying interaction screens to potent transcription factors have limited partner identification. A yeast two-hybrid screen using transcription activation-deficient mutants of the Drosophila melanogaster Hox protein Ultrabithorax IB identified an array of interacting proteins, consisting primarily of transcription factors and components of cell signaling pathways. Interactions were confirmed with wild-type Ultrabithorax (UBX) in phage display experiments and by immunoprecipitation for a subset of partners. In vivo assays demonstrated that two Ultrabithorax IB partners, Armadillo, regulated by Wingless/WNT signaling, and the homeodomain protein Aristaless, inhibit UBX-dependent haltere development from the default wing development pathway. Therefore, transcription factors and cell signaling proteins that subdivide Hox-specified tissues can both alter Hox function in vivo and interact with the corresponding Hox protein in vitro. UBX may also modulate partner function: the pupal death phenotype induced by ectopic expression of the UBX partner Hairy required the presence of UBX. Thus, Hox.transcription factor complexes may integrate a variety of positional cues, generating the specificity and versatility required for context-dependent Hox function.
Collapse
Affiliation(s)
- Sarah E Bondos
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005, USA.
| | | | | |
Collapse
|
42
|
Wesselkamper SC, Case LM, Henning LN, Borchers MT, Tichelaar JW, Mason JM, Dragin N, Medvedovic M, Sartor MA, Tomlinson CR, Leikauf GD. Gene expression changes during the development of acute lung injury: role of transforming growth factor beta. Am J Respir Crit Care Med 2005; 172:1399-411. [PMID: 16100012 PMCID: PMC2718437 DOI: 10.1164/rccm.200502-286oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE Acute lung injury can occur from multiple causes, resulting in high mortality. The pathophysiology of nickel-induced acute lung injury in mice is remarkably complex, and the molecular mechanisms are uncertain. OBJECTIVES To integrate molecular pathways and investigate the role of transforming growth factor beta (TGF-beta) in acute lung injury in mice. METHODS cDNA microarray analyses were used to identify lung gene expression changes after nickel exposure. MAPPFinder analysis of the microarray data was used to determine significantly altered molecular pathways. TGF-beta1 protein in bronchoalveolar lavage fluid, as well as the effect of inhibition of TGF-beta, was assessed in nickel-exposed mice. The effect of TGF-beta on surfactant-associated protein B (Sftpb) promoter activity was measured in mouse lung epithelial cells. MEASUREMENTS AND MAIN RESULTS Genes that decreased the most after nickel exposure play important roles in lung fluid absorption or surfactant and phospholipid synthesis, and genes that increased the most were involved in TGF-beta signaling. MAPPFinder analysis further established TGF-beta signaling to be significantly altered. TGF-beta-inducible genes involved in the regulation of extracellular matrix function and fibrinolysis were significantly increased after nickel exposure, and TGF-beta1 protein was also increased in the lavage fluid. Pharmacologic inhibition of TGF-beta attenuated nickel-induced protein in bronchoalveolar lavage. In addition, treatment with TGF-beta1 dose-dependently repressed Sftpb promoter activity in vitro, and a novel TGF-beta-responsive region in the Sftpb promoter was identified. CONCLUSIONS These data suggest that TGF-beta acts as a central mediator of acute lung injury through the alteration of several different molecular pathways.
Collapse
Affiliation(s)
- Scott C Wesselkamper
- Department of Environmental Health, P.O. Box 670056, University of Cincinnati, Cincinnati, OH 45267-0056, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Williams TM, Williams ME, Heaton JH, Gelehrter TD, Innis JW. Group 13 HOX proteins interact with the MH2 domain of R-Smads and modulate Smad transcriptional activation functions independent of HOX DNA-binding capability. Nucleic Acids Res 2005; 33:4475-84. [PMID: 16087734 PMCID: PMC1183491 DOI: 10.1093/nar/gki761] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions with co-factors provide a means by which HOX proteins exert specificity. To identify candidate protein interactors of HOXA13, we created and screened an E11.5–E12.5, distal limb bud yeast two-hybrid prey library. Among the interactors, we isolated the BMP-signaling effector Smad5, which interacted with the paralogous HOXD13 but not with HOXA11 or HOXA9, revealing unique interaction capabilities of the AbdB-like HOX proteins. Using deletion mutants, we determined that the MH2 domain of Smad5 is necessary for HOXA13 interaction. This is the first report demonstrating an interaction between HOX proteins and the MH2 domain of Smad proteins. HOXA13 and HOXD13 also bind to other BMP and TGF-β/Activin-regulated Smad proteins including Smad1 and Smad2, but not Smad4. Furthermore, HOXD13 could be co-immunoprecipitated with Smad1 from cells. Expression of HOXA13, HOXD13 or a HOXD13 homeodomain mutant (HOXD13IQN>AAA) antagonized TGF-β-stimulated transcriptional activation of the pAdtrack-3TP-Lux reporter vector in Mv1Lu cells as well as the Smad3/Smad4-activated pTRS6-E1b promoter in Hep3B cells. Finally, using mammalian one-hybrid assay, we show that transcriptional activation by a GAL4/Smad3-C-terminus fusion protein is specifically inhibited by HOXA13. Our results identify a new co-factor for HOX group 13 proteins and suggest that HOX proteins may modulate Smad-mediated transcriptional activity through protein–protein interactions without the requirement for HOX monomeric DNA-binding capability.
Collapse
Affiliation(s)
| | | | - Joanne H. Heaton
- Department of Human Genetics, University of MichiganAnn Arbor, MI, USA
| | | | - Jeffrey W. Innis
- Department of Human Genetics, University of MichiganAnn Arbor, MI, USA
- Department of Pediatrics, University of MichiganAnn Arbor, MI, USA
- To whom correspondence should be addressed. Tel: +1 734 647 3817; Fax: +1 734 763 3784;
| |
Collapse
|
44
|
Wan M, Huang J, Jhala NC, Tytler EM, Yang L, Vickers SM, Tang Y, Lu C, Wang N, Cao X. SCF(beta-TrCP1) controls Smad4 protein stability in pancreatic cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1379-92. [PMID: 15855639 PMCID: PMC1606393 DOI: 10.1016/s0002-9440(10)62356-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Smad4, also known as deleted in pancreatic carcinoma locus 4 (DPC4), is a critical co-factor in signal transduction pathways activated by transforming growth factor (TGF)-beta-related ligands that regulate cell growth and differentiation. Mutations in Smad4/DPC4 have been identified in approximately 50% of pancreatic adenocarcinomas. Here we report that SCF(beta-TrCP1), a ubiquitin (E3) ligase, is a critical determinant for Smad4 protein degradation in pancreatic cancer cells. We found that F-box protein beta-TrCP1 in this E3 ligase interacted with Smad4 and that SCF(beta-TrCP1) inhibited TGF-beta biological activity in pancreatic cancer cells by decreasing Smad4 stability. Very low Smad4 protein levels in human pancreatic ductal adenocarcinoma cells were observed by immunohistochemistry. By analyzing pancreatic tumor-derived Smad4 mutants, we found that most point-mutated Smad4 proteins, except those within or very close to a mutation cluster region, exhibited higher interaction affinity with beta-TrCP1 and significantly elevated protein ubiquitination by SCF(beta-TrCP1). Furthermore, AsPC-1 and Caco-2, two cancer cell lines harboring Smad4 point mutations, exhibited rapid Smad4 protein degradation due to the effect of SCF(beta-TrCP1). Both Smad4 levels and TGF-beta signaling were elevated by retrovirus-delivered beta-TrCP1 siRNA in pancreatic cancer cells. Therefore, inhibition of Smad4-specific E3 ligase might be a target for therapeutic intervention in pancreatic cancer.
Collapse
Affiliation(s)
- Mei Wan
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Akin ZN, Nazarali AJ. Hox genes and their candidate downstream targets in the developing central nervous system. Cell Mol Neurobiol 2005; 25:697-741. [PMID: 16075387 DOI: 10.1007/s10571-005-3971-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2003] [Accepted: 04/14/2004] [Indexed: 12/14/2022]
Abstract
1. Homeobox (Hox) genes were originally discovered in the fruit fly Drosophila, where they function through a conserved homeodomain as transcriptional regulators to control embryonic morphogenesis. Since then over 1000 homeodomain proteins have been identified in several species. In vertebrates, 39 Hox genes have been identified as homologs of the original Drosophila complex, and like their Drosophila counterparts they are organized within chromosomal clusters. Vertebrate Hox genes have also been shown to play a critical role in embryonic development as transcriptional regulators. 2. Both the Drosophila and vertebrate Hox genes have been shown to interact with various cofactors, such as the TALE homeodomain proteins, in recognition of consensus sequences within regulatory elements of their target genes. These protein-protein interactions are believed to contribute to enhancing the specificity of target gene recognition in a cell-type or tissue- dependent manner. The regulatory activity of a particular Hox protein on a specific regulatory element is highly variable and dependent on its interacting partners within the transcriptional complex. 3. In vertebrates, Hox genes display spatially restricted patterns of expression within the developing CNS, both along the anterioposterior and dorsoventral axis of the embryo. Their restricted gene expression is suggestive of a regulatory role in patterning of the CNS, as well as in cell specification. Determining the precise function of individual Hox genes in CNS morphogenesis through classical mutational analyses is complicated due to functional redundancy between Hox genes. 4. Understanding the precise mechanisms through which Hox genes mediate embryonic morphogenesis requires the identification of their downstream target genes. Although Hox genes have been implicated in the regulation of several pathways, few target genes have been shown to be under their direct regulatory control. Development of methodologies used for the isolation of target genes and for the analysis of putative targets will be beneficial in establishing the genetic pathways controlled by Hox factors. 5. Within the developing CNS various cell adhesion molecules and signaling molecules have been identified as candidate downstream target genes of Hox proteins. These targets play a role in processes such as cell migration and differentiation, and are implicated in contributing to neuronal processes such as plasticity and/or specification. Hence, Hox genes not only play a role in patterning of the CNS during early development, but may also contribute to cell specification and identity.
Collapse
Affiliation(s)
- Z N Akin
- Laboratory of Molecular Biology, College of Pharmacy and Nutrition, University of Saskatchewan, 116 Thorvaldson Building, 110 Science Place, Saskatoon, Saskatchewan, S7N 5C9, Canada
| | | |
Collapse
|
46
|
Abstract
Development of the vertebrate skeleton, a complex biological event that includes diverse processes such as formation of mesenchymal condensations at the sites of future skeletal elements, osteoblast and chondrocyte differentiation, and three dimensional patterning, is regulated by many growth factors. Bone morphogenetic proteins (BMPs), members of the TGF-beta superfamily, play a pivotal role in the signaling network and are involved in nearly all processes associated with skeletal morphogenesis. BMP signals are transduced from the plasma membrane receptors to the nucleus through both Smad pathway and non-Smad pathways, and regulated by many extracellular and intercellular proteins that interact with BMPs or components of the BMP signaling pathways. To gain a better understanding of the molecular mechanisms underlying the role of BMP in early skeletal development, it is necessary to elucidate the BMP signaling transduction pathways in chondrocytes and osteoblasts. The major objective of this review was to summarize BMP signaling pathways in the context of craniofacial, axial, and limb development. In particular, this discourse will focus on recent advances of the role of different ligands, receptors, Smads, and BMP regulators in osteoblast and chondrocyte differentiation during embryonic development.
Collapse
Affiliation(s)
- Mei Wan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
47
|
Abstract
Osteopontin (OPN) is a glyco-phosphoprotein that is expressed and secreted by numerous human cancers. OPN functions in cell adhesion, chemotaxis, macrophage-directed interleukin-10 (IL-10) suppression, stress-dependent angiogenesis, prevention of apoptosis, and anchorage-independent growth of tumor cells by regulating cell-matrix interactions and cellular signaling through binding with integrin and CD44 receptors. While constitutive expression of OPN exists in several cell types, induced expression has been detected in T-lymphocytes, epidermal cells, bone cells, macrophages, and tumor cells in remodeling processes such as inflammation, ischemia-reperfusion, bone resorption, and tumor progression. Recently, substantial evidence has linked OPN with the regulation of metastatic spread by tumor cells. However, the molecular mechanisms that define the role of OPN in tumor metastasis are incompletely understood. Transcriptional regulators that contribute to the induction of OPN expression have received significant attention as potential modulators of the OPN-mediated metastatic phenotype. The following review will discuss the molecular structure of OPN, the evidence for its functional role in tumor cell metastasis, the downstream signals that activate invasive mechanisms, and the recent reports concerning regulation of OPN transcription.
Collapse
Affiliation(s)
- Philip Y Wai
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
48
|
Renault MA, Jalvy S, Potier M, Belloc I, Genot E, Dekker LV, Desgranges C, Gadeau AP. UTP induces osteopontin expression through a coordinate action of NFkappaB, activator protein-1, and upstream stimulatory factor in arterial smooth muscle cells. J Biol Chem 2004; 280:2708-13. [PMID: 15557322 DOI: 10.1074/jbc.m411786200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Osteopontin (OPN) is an important chemokinetic agent for several cell types. Our earlier studies have shown that its expression is essential for uridine triphosphate (UTP)-mediated migration of vascular smooth muscle cells. We demonstrated previously that the activation of an AP-1 binding site located 76 bp upstream of the transcription start in the rat OPN promoter is involved in the induction of OPN expression. In this work, using a luciferase promoter deletion assay, we identified a new region of the rat OPN promoter (-1837 to -1757) that is responsive to UTP. This region contains an NFkappaB site located at -1800 and an Ebox located at -1768. Supershift electrophoretic mobility shift assay and chromatin immunoprecipitation assays identified NFkappaB and USF-1/USF-2 as the DNA binding proteins induced by UTP, respectively, for these two sites. Using dominant negative mutants of IkappaB kinase and USF transcription factors, we confirmed that NFkappaB and USF-1/USF-2 are involved in the UTP-mediated expression of OPN. Using a pharmacological approach, we demonstrated that USF proteins are regulated by the extracellular signal-regulated kinase (ERK)1/2 pathway, just as the earlier discovered AP-1 complex, whereas NFkappaB is up-regulated through PKCdelta signals. Finally, our work suggests that the UTP-stimulated OPN expression involves a coordinate regulation of PKCdelta-NFkappaB, ERK1/2-USF, and ERK1/2/NAD(P)H oxidase AP-1 signaling pathways.
Collapse
|
49
|
Subramanian SV, Polikandriotis JA, Kelm RJ, David JJ, Orosz CG, Strauch AR. Induction of vascular smooth muscle alpha-actin gene transcription in transforming growth factor beta1-activated myofibroblasts mediated by dynamic interplay between the Pur repressor proteins and Sp1/Smad coactivators. Mol Biol Cell 2004; 15:4532-43. [PMID: 15282343 PMCID: PMC519147 DOI: 10.1091/mbc.e04-04-0348] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The mouse vascular smooth muscle alpha-actin (SMA) gene enhancer is activated in fibroblasts by transforming growth factor beta1 (TGFbeta1), a potent mediator of myofibroblast differentiation and wound healing. The SMA enhancer contains tandem sites for the Sp1 transcriptional activator protein and Puralpha and beta repressor proteins. We have examined dynamic interplay between these divergent proteins to identify checkpoints for possible control of myofibroblast differentiation during chronic inflammatory disease. A novel element in the SMA enhancer named SPUR was responsible for both basal and TGFbeta1-dependent transcriptional activation in fibroblasts and capable of binding Sp1 and Pur proteins. A novel Sp1:Pur:SPUR complex was dissociated when SMA enhancer activity was increased by TGFbeta1 or Smad protein overexpression. Physical association of Pur proteins with Smad2/3 was observed as was binding of Smads to an upstream enhancer region that undergoes DNA duplex unwinding in TGFbeta1-activated myofibroblasts. Purbeta repression of the SMA enhancer could not be relieved by TGFbeta1, whereas repression mediated by Puralpha was partially rescued by TGFbeta1 or overexpression of Smad proteins. Interplay between Pur repressor isoforms and Sp1 and Smad coactivators may regulate SMA enhancer output in TGFbeta1-activated myofibroblasts during episodes of wound repair and tissue remodeling.
Collapse
Affiliation(s)
- Sukanya V Subramanian
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
50
|
Aherrahrou Z, Axtner SB, Kaczmarek PM, Jurat A, Korff S, Doehring LC, Weichenhan D, Katus HA, Ivandic BT. A locus on chromosome 7 determines dramatic up-regulation of osteopontin in dystrophic cardiac calcification in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1379-87. [PMID: 15039225 PMCID: PMC1615353 DOI: 10.1016/s0002-9440(10)63224-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Calcification of necrotic tissue is frequently observed in chronic inflammation and atherosclerosis. A similar response of myocardium to injury, referred to as dystrophic cardiac calcinosis (DCC), occurs in certain inbred strains of mice. We now examined a putative inhibitor of calcification, osteopontin, in DCC after transdiaphragmal myocardial freeze-thaw injury. Strong osteopontin expression was found co-localizing with calcification in DCC-susceptible strain C3H/HeNCrlBr, which exhibited low osteopontin plasma concentrations otherwise. Osteopontin mRNA induction was 20-fold higher than in resistant strain C57BL/6NCrlBr, which exhibited fibrous lesions without calcification and little osteopontin expression. Sequence analysis identified several polymorphisms in calcium-binding and phosphorylation sites in osteopontin cDNA. Their potential relevance for DCC was tested in congenic mice, which shared the osteopontin locus with C57BL/6NCrlBr, but retained a chromosomal segment from C3H/HeNCrlBr on proximal chromosome 7. These mice exhibited strong osteopontin expression and DCC comparable to C3H/HeNCrlBr suggesting that a trans-activator of osteopontin transcription residing on chromosome 7 and not the osteopontin gene on chromosome 5 was responsible for the genetic differences in osteopontin expression. A known osteopontin activator encoded by a gene on chromosome 7 is the transforming growth factor-beta1, which was more induced (3.5x) in C3H/HeNCrlBr than in C57BL/6NCrlBr mice.
Collapse
|