1
|
Xiong D, Tong CS, Wu M. A molecular systems perspective on calcium oscillations beyond ion fluxes. Curr Opin Cell Biol 2025; 94:102523. [PMID: 40311263 DOI: 10.1016/j.ceb.2025.102523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Calcium (Ca2+) oscillations, marked by periodic fluctuations in cytosolic Ca2+ levels, are a universal feature of both excitable and non-excitable cells, regulating key functions like immune responses, neuronal activity and oocyte activation. Despite significant progress over the past few decades in identifying the molecular toolkits involved in Ca2+ mobilization, fundamental questions remain unresolved: How do Ca2+oscillations arise? In dynamical systems, oscillations arise as closed-loop trajectories in phase space, known as limit cycles. In this framework, [Ca2+] is the variable that oscillates along the limit cycle. Is [Ca2+] also the control parameter that defines the system's stability? Understanding how oscillations arise and how instability is controlled are essential for determining what these oscillations encode. This review revisits classic categorizations of Ca2+ oscillation models, focusing on the minimal mathematical models, their assumptions and gaps linking models with experimental data. We examine historical arguments in light of recent discoveries of plasma membrane lipid oscillations in non-excitable cells. While growing evidence support the pivotal role of lipid signaling in regulating Ca2+ dynamics, they mostly focused on the upstream role of signaling in Ca2+ mobilization, rather than viewing membrane-dependent signal transduction as the core control loop that is responsible for oscillatory Ca2+ dynamics. Here we summarize recent molecular studies of phosphoinositide signaling in modulating Ca2+ dynamics, by considering a broader chemical perspective as essential for understanding Ca2+ oscillations beyond ion fluxes.
Collapse
Affiliation(s)
- Ding Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Chee San Tong
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8002, USA
| | - Min Wu
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8002, USA.
| |
Collapse
|
2
|
Peng V, Trsan T, Sudan R, Bhattarai B, Cortez VS, Molgora M, Vacher J, Colonna M. Inositol phosphatase INPP4B sustains ILC1s and intratumoral NK cells through an AKT-driven pathway. J Exp Med 2024; 221:e20230124. [PMID: 38197946 PMCID: PMC10783437 DOI: 10.1084/jem.20230124] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 11/17/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population of lymphocytes that coordinate early immune responses and maintain tissue homeostasis. Type 1 innate immune responses are mediated by natural killer (NK) cells and group 1 ILCs (ILC1s). Despite their shared features, NK cells and ILC1s display profound differences among various tissue microenvironments. Here, we identify the inositol polyphosphatase INPP4B as a hallmark feature of tissue-resident ILC1s and intratumoral NK cells using an scRNA-seq atlas of tissue-associated and circulating NK/ILC1s. Conditional deletion of Inpp4b in ILC1s and NK cells reveals that it is necessary for the homeostasis of tissue-resident ILC1s but not circulating NK cells at steady-state. Inpp4b-deficient cells display increased rates of apoptosis and reduced activation of the prosurvival molecule AKT. Furthermore, expression of Inpp4b by NK/ILC1s is necessary for their presence in the intratumoral environment, and lack of Inpp4b impairs antitumor immunity. These findings highlight INPP4B as a novel regulator of tissue residency and antitumor function in ILC1s and NK cells.
Collapse
Affiliation(s)
- Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Raki Sudan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bishan Bhattarai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Victor S. Cortez
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal, Montréal, Canada
- Département de Médecine, Université de Montréal, Montréal, Canada
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Dinicola S, Unfer V, Soulage CO, Margarita Yap-Garcia MI, Bevilacqua A, Benvenga S, Barbaro D, Wdowiak A, Nordio M, Dewailly D, Appetecchia M, Aragona C, Bezerra Espinola MS, Bizzarri M, Cavalli P, Colao A, D’Anna R, Vazquez-Levin MH, Marin IH, Kamenov Z, Laganà AS, Monastra G, Oliva MM, Özay AC, Pintaudi B, Porcaro G, Pustotina O, Pkhaladze L, Prapas N, Roseff S, Salehpour S, Stringaro A, Tugushev M, Unfer V, Vucenik I, Facchinetti F. <sc>d</sc>-Chiro-Inositol in Clinical Practice: A Perspective from the Experts Group on Inositol in Basic and Clinical Research (EGOI). Gynecol Obstet Invest 2024; 89:284-294. [PMID: 38373412 PMCID: PMC11309080 DOI: 10.1159/000536081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND d-Chiro-inositol is a natural molecule that, in association with its well-studied isomer myo-inositol, may play a role in treating various metabolic and gynecological disorders. OBJECTIVES This perspective seeks to explore the mechanisms and functions of d-chiro-inositol, laying the foundations to discuss its use in clinical practice, across dysmetabolism, obesity, and hormonal dysregulation. METHODS A narrative review of all the relevant papers known to the authors was conducted. OUTCOME d-Chiro-inositol acts through a variety of mechanisms, acting as an insulin sensitizer, inhibiting the transcription of aromatase, in addition to modulating white adipose tissue/brown adipose tissue transdifferentiation. These different modes of action have potential applications in a variety of therapeutic fields, including PCOS, dysmetabolism, obesity, hypoestrogenic/hyperandrogenic disorders, and bone health. CONCLUSIONS d-Chiro-inositol mode of action has been studied in detail in recent years, resulting in a clear differentiation between d-chiro-inositol and its isomer myo-inositol. The insulin-sensitizing activities of d-chiro-inositol are well understood; however, its potential applications in other fields, in particular obesity and hyperestrogenic/hypoandrogenic disorders in men and women, represent promising avenues of research that require further clinical study.
Collapse
Affiliation(s)
- Simona Dinicola
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Systems Biology Group Lab, Rome, Italy
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- UniCamillus – Saint Camillus International University of Health Sciences, Rome, Italy
| | - Christophe O. Soulage
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- INSERM U1060, INSA de Lyon, University of Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Maria Isidora Margarita Yap-Garcia
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- St. Luke’s Medical Center College of Medicine, William H. Quasha Memorial, Quezon, Philippines
| | - Arturo Bevilacqua
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Dynamic, Clinical Psychology and Health, Sapienza University of Rome, Rome, Italy
| | - Salvatore Benvenga
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Daniele Barbaro
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Director of U.O. Endocrinology in Livorno Hospital, Livorno, Italy
| | - Artur Wdowiak
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Faculty of Medicine and Dentistry, Medical University of Lublin, Lublin, Poland
| | - Maurizio Nordio
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- A.S.L. RMF, Civitavecchia, Italy
| | - Didier Dewailly
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Faculty of Medicine Henri Warembourg, University of Lille, Lille Cedex, France
| | - Marialuisa Appetecchia
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Oncological Endocrinology Unit, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Cesare Aragona
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Systems Biology Group Lab, Rome, Italy
| | - Maria Salomè Bezerra Espinola
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Systems Biology Group Lab, Rome, Italy
| | - Mariano Bizzarri
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Systems Biology Group Lab, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Pietro Cavalli
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Annamaria Colao
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Italian Society of Endocrinology, Federico II University of Naples, Naples, Italy
| | - Rosario D’Anna
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Mónica Hebe Vazquez-Levin
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- National Council of Scientific and Technical Research, Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina
| | - Imelda Hernàndez Marin
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Human Reproduction Department, Hospital Juárez de México, and Universidad Nacional Autónoma de México (UNAM), México, Mexico
| | - Zdravko Kamenov
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Internal Medicine, University Hospital “Alexandrovska”, Clinic of Endocrinology and Metabolism, Medical University, Sofia, Bulgaria
| | - Antonio Simone Laganà
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giovanni Monastra
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
| | - Mario Montanino Oliva
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Obstetrics and Gynecology, Santo Spirito Hospital, Rome, Italy
| | - Ali Cenk Özay
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Obstetrics and Gynecology, Faculty of Medicine, Cyprus International University, Nicosia, Cyprus
| | - Basilio Pintaudi
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giuseppina Porcaro
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Women's Health Centre, USL UMBRIA 2, Terni, Italy
| | - Olga Pustotina
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Obstetrics and Gynecology with Reproductive Medicine, F.I. Inozemtsev Academy of Medical Education, Saint Petersburg, Russia
| | - Lali Pkhaladze
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Zhordania and Khomasuridze Institute of Reproductology, Tbilisi, Georgia
| | - Nikos Prapas
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Third Department of OB-GYNAE, Aristotle University of Thessaloniki, and IVF Laboratory, IAKENTRO Fertility Centre, Thessaloniki, Greece
| | - Scott Roseff
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Reproductive Endocrinology and Infertility, South Florida Institute for Reproductive Medicine (IVFMD), Jupiter, FL, USA
| | - Saghar Salehpour
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Annarita Stringaro
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- National Center for Drug Research and Evaluation, Italian National Institute of Health, Rome, Italy
| | - Marat Tugushev
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Reproductive Medicine, Clinical Embryology and Genetics of Samara State Medical University, Samara, Russia
| | - Virginia Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- A.G.Un.Co. Obstetrics and Gynecology Center, Rome, Italy
| | - Ivana Vucenik
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- Department of Medical and Research Technology and Pathology, University of Maryland School of Medicine in Baltimore, Baltimore, MD, USA
| | - Fabio Facchinetti
- The Experts Group on Inositol in Basic and Clinical Research (EGOI) Rome, Italy
- University of Modena and Reggio Emilia, Modena, Italy
- President Italian Society of Perinatal Medicine (SIMP), Modena, Italy
| |
Collapse
|
4
|
Zhang M, Ceyhan Y, Mei S, Hirz T, Sykes DB, Agoulnik IU. Regulation of EZH2 Expression by INPP4B in Normal Prostate and Primary Prostate Cancer. Cancers (Basel) 2023; 15:5418. [PMID: 38001678 PMCID: PMC10670027 DOI: 10.3390/cancers15225418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
The phosphatases INPP4B and PTEN are tumor suppressors that are lost in nearly half of advanced metastatic cancers. The loss of PTEN in prostate epithelium initially leads to an upregulation of several tumor suppressors that slow the progression of prostate cancer in mouse models. We tested whether the loss of INPP4B elicits a similar compensatory response in prostate tissue and whether this response is distinct from the one caused by the loss of PTEN. Knockdown of INPP4B but not PTEN in human prostate cancer cell lines caused a decrease in EZH2 expression. In Inpp4b-/- mouse prostate epithelium, EZH2 levels were decreased, as were methylation levels of histone H3. In contrast, Ezh2 levels were increased in the prostates of Pten-/- male mice. Contrary to PTEN, there was a positive correlation between INPP4B and EZH2 expression in normal human prostates and early-stage prostate tumors. Analysis of single-cell transcriptomic data demonstrated that a subset of EZH2-positive cells expresses INPP4B or PTEN, but rarely both, consistent with their opposing correlation with EZH2 expression. Unlike PTEN, INPP4B did not affect the levels of SMAD4 protein expression or Pml mRNA expression. Like PTEN, p53 protein expression and phosphorylation of Akt in Inpp4b-/- murine prostates were elevated. Taken together, the loss of INPP4B in the prostate leads to overlapping and distinct changes in tumor suppressor and oncogenic downstream signaling.
Collapse
Affiliation(s)
- Manqi Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA;
| | - Yasemin Ceyhan
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Shenglin Mei
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Taghreed Hirz
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Irina U. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami, FL 33199, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Lacombe J, Guo K, Bonneau J, Faubert D, Gioanni F, Vivoli A, Muir SM, Hezzaz S, Poitout V, Ferron M. Vitamin K-dependent carboxylation regulates Ca 2+ flux and adaptation to metabolic stress in β cells. Cell Rep 2023; 42:112500. [PMID: 37171959 DOI: 10.1016/j.celrep.2023.112500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 02/24/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
Vitamin K is a micronutrient necessary for γ-carboxylation of glutamic acids. This post-translational modification occurs in the endoplasmic reticulum (ER) and affects secreted proteins. Recent clinical studies implicate vitamin K in the pathophysiology of diabetes, but the underlying molecular mechanism remains unknown. Here, we show that mouse β cells lacking γ-carboxylation fail to adapt their insulin secretion in the context of age-related insulin resistance or diet-induced β cell stress. In human islets, γ-carboxylase expression positively correlates with improved insulin secretion in response to glucose. We identify endoplasmic reticulum Gla protein (ERGP) as a γ-carboxylated ER-resident Ca2+-binding protein expressed in β cells. Mechanistically, γ-carboxylation of ERGP protects cells against Ca2+ overfilling by diminishing STIM1 and Orai1 interaction and restraining store-operated Ca2+ entry. These results reveal a critical role of vitamin K-dependent carboxylation in regulation of Ca2+ flux in β cells and in their capacity to adapt to metabolic stress.
Collapse
Affiliation(s)
- Julie Lacombe
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada.
| | - Kevin Guo
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Jessica Bonneau
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Denis Faubert
- Mass Spectrometry and Proteomics Platform, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Florian Gioanni
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Alexis Vivoli
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Sarah M Muir
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Soraya Hezzaz
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
6
|
Tumor Suppressor Role of INPP4B in Chemoresistant Retinoblastoma. JOURNAL OF ONCOLOGY 2023; 2023:2270097. [PMID: 36993823 PMCID: PMC10042642 DOI: 10.1155/2023/2270097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023]
Abstract
The chemotherapy of retinoblastoma (RB), a malignant ocular childhood disease, is often limited by the development of resistance against commonly used drugs. We identified inositol polyphosphate 4-phosphatase type II (INPP4B) as a differentially regulated gene in etoposide-resistant RB cell lines, potentially involved in the development of RB resistances. INPP4B is controversially discussed as a tumor suppressor and an oncogenic driver in various cancers, but its role in retinoblastoma in general and chemoresistant RB in particular is yet unknown. In the study presented, we investigated the expression of INPP4B in RB cell lines and patients and analyzed the effect of INPP4B overexpression on etoposide resistant RB cell growth in vitro and in vivo. INPP4B mRNA levels were significantly downregulated in RB cells lines compared to the healthy human retina, with even lower expression levels in etoposide-resistant compared to the sensitive cell lines. Besides, a significant increase in INPP4B expression was observed in chemotherapy-treated RB tumor patient samples compared to untreated tumors. INPP4B overexpression in etoposide-resistant RB cells resulted in a significant reduction in cell viability with reduced growth, proliferation, anchorage-independent growth, and in ovo tumor formation. Caspase-3/7-mediated apoptosis was concomitantly increased, suggesting a tumor suppressive role of INPP4B in chemoresistant RB cells. No changes in AKT signaling were discernible, but p-SGK3 levels increased following INPP4B overexpression, indicating a potential regulation of SGK3 signaling in etoposide-resistant RB cells. RNAseq analysis of INPP4B overexpressing, etoposide-resistant RB cell lines revealed differentially regulated genes involved in cancer progression, mirroring observed in vitro and in vivo effects of INPP4B overexpression and strengthening INPP4B’s importance for cell growth control and tumorigenicity.
Collapse
|
7
|
Gao ZR, Feng YZ, Zhao YQ, Zhao J, Zhou YH, Ye Q, Chen Y, Tan L, Zhang SH, Feng Y, Hu J, Ou-Yang ZY, Dusenge MA, Guo Y. Traditional Chinese medicine promotes bone regeneration in bone tissue engineering. Chin Med 2022; 17:86. [PMID: 35858928 PMCID: PMC9297608 DOI: 10.1186/s13020-022-00640-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/30/2022] [Indexed: 11/10/2022] Open
Abstract
Bone tissue engineering (BTE) is a promising method for the repair of difficult-to-heal bone tissue damage by providing three-dimensional structures for cell attachment, proliferation, and differentiation. Traditional Chinese medicine (TCM) has been introduced as an effective global medical program by the World Health Organization, comprising intricate components, and promoting bone regeneration by regulating multiple mechanisms and targets. This study outlines the potential therapeutic capabilities of TCM combined with BTE in bone regeneration. The effective active components promoting bone regeneration can be generally divided into flavonoids, alkaloids, glycosides, terpenoids, and polyphenols, among others. The chemical structures of the monomers, their sources, efficacy, and mechanisms are described. We summarize the use of compounds and medicinal parts of TCM to stimulate bone regeneration. Finally, the limitations and prospects of applying TCM in BTE are introduced, providing a direction for further development of novel and potential TCM.
Collapse
Affiliation(s)
- Zheng-Rong Gao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yun-Zhi Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ya-Qiong Zhao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Jie Zhao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ying-Hui Zhou
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Ye
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yun Chen
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Li Tan
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Shao-Hui Zhang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yao Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Jing Hu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ze-Yue Ou-Yang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Marie Aimee Dusenge
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yue Guo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Mutabaruka MS, Pata M, Vacher J. A Foxo1-Klf2-S1pr1-Gnai1-Rac1 signaling axis is a critical mediator of Ostm1 regulatory network in T lymphopoiesis. iScience 2022; 25:104160. [PMID: 35434560 PMCID: PMC9010627 DOI: 10.1016/j.isci.2022.104160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/23/2022] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
Ostm1 mutations cause the severe form of osteopetrosis with bone marrow deficiency in humans and mice, yet a role in T cell ontogeny remains to be determined. Herein, we show that thymi of the Ostm1-null mice (gl/gl) from P8-to-P15 become markedly hypocellular with disturbed architecture. Analysis of gl/gl early T cell program determined a major decrease of 3-fold in bone marrow common lymphoid precursors (CLP), 35-fold in early thymic precursors (ETPs) and 100-fold in T cell double positive subpopulations. Ostm1 ablation in T cell double negative (DN) also appears to induce fast-paced differentiation kinetics with a transitory intermediate CD44+CD25int subpopulation. Transgenic targeting Ostm1 expression from the gl/gl DN1 population partially rescued T cell subpopulations from ETP onwards and normalized the accelerated DN differentiation, indicating a cell-autonomous role for Ostm1. Transcriptome of early DN1 population identified an Ostm1 crosstalk with a Foxo1-Klf2-S1pr1-Gnai1-Rac1 signaling axis. Our findings establish that Ostm1 is an essential regulator of T cell ontogeny. Loss of Ostm1 causes severe thymus hypocellularity Ostm1 is a modulator of the T cell differentiation program from the CLPs onwards Targeted CD2-Ostm1 in Ostm1 null mice leads to partial rescue of DN differentiation Ostm1 null DN1 transcriptome identifies a Foxo1-Klf2-S1pr1-Gnai1-Rac1 signaling axis
Collapse
Affiliation(s)
- Marie S Mutabaruka
- Institut de Recherches Cliniques de Montréal, 110 West Pins Avenue, Montréal, QC H2W 1R7, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC H3A 1A3, Canada
| | - Monica Pata
- Institut de Recherches Cliniques de Montréal, 110 West Pins Avenue, Montréal, QC H2W 1R7, Canada
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal, 110 West Pins Avenue, Montréal, QC H2W 1R7, Canada.,Département de Médecine, Université de Montréal, Montréal, QC H3T 3J7, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC H3A 1A3, Canada
| |
Collapse
|
9
|
Zhang H, Wu Z. The generalized Fisher's combination and accurate p-value calculation under dependence. Biometrics 2022. [PMID: 35178716 DOI: 10.1111/biom.13634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
Abstract
Combining dependent tests of significance has broad applications but the related p-value calculation is challenging. For Fisher's combination test, current p-value calculation methods (e.g., Brown's approximation) tend to inflate the type I error rate when the desired significance level is substantially less than 0.05. The problem could lead to significant false discoveries in big data analyses. This paper provides two main contributions. First, it presents a general family of Fisher type statistics, referred to as the GFisher, which covers many classic statistics, such as Fisher's combination, Good's statistic, Lancaster's statistic, weighted Z-score combination, etc. The GFisher allows a flexible weighting scheme, as well as an omnibus procedure that automatically adapts proper weights and the statistic-defining parameters to a given data. Second, the paper presents several new p-value calculation methods based on two novel ideas: moment-ratio matching and joint-distribution surrogating. Systematic simulations show that the new calculation methods are more accurate under multivariate Gaussian, and more robust under the generalized linear model and the multivariate t-distribution. The applications of the GFisher and the new p-value calculation methods are demonstrated by a gene-based SNP-set association study. Relevant computation has been implemented to an R package GFisher available on the Comprehensive R Archive Network. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hong Zhang
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Rahway, New Jersey, U.S.A
| | - Zheyang Wu
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, Massachusetts, U.S.A
| |
Collapse
|
10
|
Hamila SA, Ooms LM, Rodgers SJ, Mitchell CA. The INPP4B paradox: Like PTEN, but different. Adv Biol Regul 2021; 82:100817. [PMID: 34216856 DOI: 10.1016/j.jbior.2021.100817] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/28/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
Cancer is a complex and heterogeneous disease marked by the dysregulation of cancer driver genes historically classified as oncogenes or tumour suppressors according to their ability to promote or inhibit tumour development and growth, respectively. Certain genes display both oncogenic and tumour suppressor functions depending on the biological context, and as such have been termed dual-role cancer driver genes. However, because of their context-dependent behaviour, the tumourigenic mechanism of many dual-role genes is elusive and remains a significant knowledge gap in our effort to understand and treat cancer. Inositol polyphosphate 4-phosphatase type II (INPP4B) is an emerging dual-role cancer driver gene, primarily known for its role as a negative regulator of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway. In response to growth factor stimulation, class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane. PtdIns(3,4,5)P3 can be hydrolysed by inositol polyphosphate 5-phosphatases to generate PtdIns(3,4)P2, which, together with PtdIns(3,4,5)P3, facilitates the activation of AKT to promote cell proliferation, survival, migration, and metabolism. Phosphatase and tensin homology on chromosome 10 (PTEN) and INPP4B are dual-specificity phosphatases that hydrolyse PtdIns(3,4,5)P3 and PtdIns(3,4)P2, respectively, and thus negatively regulate PI3K/AKT signalling. PTEN is a bona fide tumour suppressor that is frequently lost in human tumours. INPP4B was initially characterised as a tumour suppressor akin to PTEN, and has been implicated as such in a number of cancers, including prostate, thyroid, and basal-like breast cancers. However, evidence has since emerged revealing INPP4B as a paradoxical oncogene in several malignancies, with increased INPP4B expression reported in AML, melanoma and colon cancers among others. Although the tumour suppressive function of INPP4B has been mostly ascribed to its ability to negatively regulate PI3K/AKT signalling, its oncogenic function remains less clear, with proposed mechanisms including promotion of PtdIns(3)P-dependent SGK3 signalling, inhibition of PTEN-dependent AKT activation, and enhancing DNA repair mechanisms to confer chemoresistance. Nevertheless, research is ongoing to identify the factors that dictate the tumourigenic output of INPP4B in different human cancers. In this review we discuss the dualistic role that INPP4B plays in the context of cancer development, progression and treatment, drawing comparisons to PTEN to explore how their similarities and, importantly, their differences may account for their diverging roles in tumourigenesis.
Collapse
Affiliation(s)
- Sabryn A Hamila
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Lisa M Ooms
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Samuel J Rodgers
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
11
|
The Aqueous Extract of Eucommia Leaves Promotes Proliferation, Differentiation, and Mineralization of Osteoblast-Like MC3T3-E1 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:3641317. [PMID: 34249129 PMCID: PMC8238580 DOI: 10.1155/2021/3641317] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023]
Abstract
Eucommia leaves are dry leaves of Eucommia ulmoides which have long been considered as a functional health food for the treatment of hypertension, hypercholesterolemia, fatty liver, and osteoporosis. With the recent development of Chinese medicine, Eucommia leaves are widely used for tonifying the kidneys and strengthening bone. However, the specific molecular mechanism of Eucommia leaves for strengthening bone remains largely unknown. Osteoblasts are the main functional cells of bone formation; thus, it is essential to study the effect of Eucommia leaves on osteoblasts to better understand their mechanism of action. In the present study, we prepared an aqueous extract of Eucommia leaves (ELAE) and determined its content by high-performance liquid chromatography (HPLC). The effects of ELAE on MC3T3-E1 cells were investigated by CCK-8 assay, alkaline phosphatase (ALP), and Alizarin red S staining assays, combined with RNA sequencing (RNA-seq) and qRT-PCR validation. We demonstrated that ELAE had a significant promoting effect on the proliferation of MC3T3-E1 cells and significantly enhanced extracellular matrix synthesis and mineralization, which were achieved by regulating various functional genes and related signaling pathways. ELAE significantly increased the expression level of genes promoting cell proliferation, such as Rpl10a, Adnp, Pex1, Inpp4a, Frat2, and Pcdhga1, and reduced the expression level of genes inhibiting cell proliferation, such as Npm1, Eif3e, Cbx3, Psmc6, Fgf7, Fxr1, Ddx3x, Mbnl1, and Cdc27. In addition, ELAE increased the expression level of gene markers in osteoblasts, such as Col5a2, Ubap2l, Dkk3, Foxm1, Col16a1, Col12a1, Usp7, Col4a6, Runx2, Sox4, and Bmp4. Taken together, our results suggest that ELAE could promote osteoblast proliferation, differentiation, and mineralization and prevent osteoblast apoptosis. These findings not only increase our understanding of ELAE on the regulation of bone development but also provide a possible strategy to further study the prevention and treatment of osteogenic related diseases by ELAE.
Collapse
|
12
|
Zhang M, Ceyhan Y, Kaftanovskaya EM, Vasquez JL, Vacher J, Knop FK, Nathanson L, Agoulnik AI, Ittmann MM, Agoulnik IU. INPP4B protects from metabolic syndrome and associated disorders. Commun Biol 2021; 4:416. [PMID: 33772116 PMCID: PMC7998001 DOI: 10.1038/s42003-021-01940-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
A high fat diet and obesity have been linked to the development of metabolic dysfunction and the promotion of multiple cancers. The causative cellular signals are multifactorial and not yet completely understood. In this report, we show that Inositol Polyphosphate-4-Phosphatase Type II B (INPP4B) signaling protects mice from diet-induced metabolic dysfunction. INPP4B suppresses AKT and PKC signaling in the liver thereby improving insulin sensitivity. INPP4B loss results in the proteolytic cleavage and activation of a key regulator in de novo lipogenesis and lipid storage, SREBP1. In mice fed with the high fat diet, SREBP1 increases expression and activity of PPARG and other lipogenic pathways, leading to obesity and non-alcoholic fatty liver disease (NAFLD). Inpp4b-/- male mice have reduced energy expenditure and respiratory exchange ratio leading to increased adiposity and insulin resistance. When treated with high fat diet, Inpp4b-/- males develop type II diabetes and inflammation of adipose tissue and prostate. In turn, inflammation drives the development of high-grade prostatic intraepithelial neoplasia (PIN). Thus, INPP4B plays a crucial role in maintenance of overall metabolic health and protects from prostate neoplasms associated with metabolic dysfunction.
Collapse
Affiliation(s)
- Manqi Zhang
- Department of Medicine, Duke University, Durham, NC, USA
| | - Yasemin Ceyhan
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Elena M Kaftanovskaya
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Judy L Vasquez
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Jean Vacher
- Department of Medicine, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, USA
| | - Irina U Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Pata M, Yousefi Behzadi P, Vacher J. Expression pattern of the V5-Ostm1 protein in bacterial artificial chromosome transgenic mice. Genesis 2021; 59:e23409. [PMID: 33484096 DOI: 10.1002/dvg.23409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 11/08/2022]
Abstract
Mutations in the osteopetrotic transmembrane protein 1 (Ostm1) gene are responsible for the most severe form of autosomal recessive osteopetrosis both in humans and in the gray lethal (gl/gl) mouse. This defect leads to increased bone mass with bone marrow occlusion and hematopoietic defects. To establish the expression profile of the mouse Ostm1 protein in vivo, homologous recombination in bacteria was designed to generate a V5-Ostm1 bacterial artificial chromosome (BAC) that was subsequently integrated in the mouse genome. Tissue expression of the transgene V5-Ostm1 RNA and protein in transgenic mice follow the endogenous expression profile. Immunohistochemistry analysis demonstrated expression in neuronal populations from central and peripheral nervous system and defined a unique cellular expression pattern. Importantly, together with appropriate protein post-translational modification, in vivo rescue of the osteopetrotic bone gl/gl phenotype in BAC V5-Ostm1 gl/gl mice is consistent with the expression of a fully functional and active protein. These mice represent a unique tool to unravel novel Ostm1 functions in individual tissue and neuronal cell populations and the V5-Ostm1 transgene represents an easy visual marker to monitor the expression of Ostm1 in vitro and in vivo.
Collapse
Affiliation(s)
- Monica Pata
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Pardis Yousefi Behzadi
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
14
|
Vacher J, Bruccoleri M, Pata M. Ostm1 from Mouse to Human: Insights into Osteoclast Maturation. Int J Mol Sci 2020; 21:ijms21165600. [PMID: 32764302 PMCID: PMC7460669 DOI: 10.3390/ijms21165600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
The maintenance of bone mass is a dynamic process that requires a strict balance between bone formation and resorption. Bone formation is controlled by osteoblasts, while osteoclasts are responsible for resorption of the bone matrix. The opposite functions of these cell types have to be tightly regulated not only during normal bone development, but also during adult life, to maintain serum calcium homeostasis and sustain bone integrity to prevent bone fractures. Disruption of the control of bone synthesis or resorption can lead to an over accumulation of bone tissue in osteopetrosis or conversely to a net depletion of the bone mass in osteoporosis. Moreover, high levels of bone resorption with focal bone formation can cause Paget’s disease. Here, we summarize the steps toward isolation and characterization of the osteopetrosis associated trans-membrane protein 1 (Ostm1) gene and protein, essential for proper osteoclast maturation, and responsible when mutated for the most severe form of osteopetrosis in mice and humans.
Collapse
Affiliation(s)
- Jean Vacher
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A3, Canada
- Correspondence:
| | - Michael Bruccoleri
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
| | - Monica Pata
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
| |
Collapse
|
15
|
Ceyhan Y, Zhang M, Guo J, Sandoval CG, Vacher J, Kaftanovskaya EM, Agoulnik AI, Agoulnik IU. Deletion of inositol polyphosphate 4-phosphatase type-II B affects spermatogenesis in mice. PLoS One 2020; 15:e0233163. [PMID: 32413098 PMCID: PMC7228085 DOI: 10.1371/journal.pone.0233163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Inositol polyphosphate-4-phosphatase type II (INPP4B) is a dual-specificity phosphatase that acts as a tumor suppressor in multiple cancers. INPP4B dephosphorylates phospholipids at the 4th position of the inositol ring and inhibits AKT and PKC signaling by hydrolyzing of PI(3,4)P2 and PI(4,5)P2, respectively. INPP4B protein phosphatase targets include phospho-tyrosines on Akt and phospho-serine and phospho-threonine on PTEN. INPP4B is highly expressed in testes, suggesting its role in testes development and physiology. The objective of this study was to determine whether Inpp4b deletion impacts testicular function in mice. In testis, Inpp4b expression was the highest in postmeiotic germ cells in both mice and men. The testes of Inpp4b knockout male mice were significantly smaller compared to the testes of wild-type (WT) males. Inpp4b-/- males produced fewer mature sperm cells compared to WT, and this difference increased with age and high fat diet (HFD). Reduction in early steroidogenic enzymes and luteinizing hormone (LH) receptor gene expression was detected, although androgen receptor (AR) protein level was similar in WT and Inpp4b-/- testes. Germ cell apoptosis was significantly increased in the knockout mice, while expression of meiotic marker γH2A.X was decreased. Our data demonstrate that INPP4B plays a role in maintenance of male germ cell differentiation and protects testis functions against deleterious effects of aging and high fat diet.
Collapse
Affiliation(s)
- Yasemin Ceyhan
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Manqi Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, United States of America
| | - Jingtao Guo
- Department of Oncological Sciences and Huntsman Cancer Institute, Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Surgery (Andrology/Urology), Center for Reconstructive Urology and Men’s Health, University of Utah Health Sciences Center, Salt Lake City, UT, United States of America
| | - Carlos G. Sandoval
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Jean Vacher
- Department of Medicine, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Elena M. Kaftanovskaya
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Alexander I. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
- Biomolecular Science Institute, Florida International University, Miami, FL, United States of America
| | - Irina U. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
- Biomolecular Science Institute, Florida International University, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
16
|
Mangialardi EM, Chen K, Salmon B, Vacher J, Salmena L. Investigating the duality of Inpp4b function in the cellular transformation of mouse fibroblasts. Oncotarget 2019; 10:6378-6390. [PMID: 31695845 PMCID: PMC6824866 DOI: 10.18632/oncotarget.27293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/19/2019] [Indexed: 11/25/2022] Open
Abstract
Inositol Polyphosphate 4-Phosphatase, Type II (INPP4B) is a tumour suppressor in breast, ovarian, prostate, thyroid and other cancers, attributed to its ability to reduce oncogenic Akt-signaling. However, emerging studies show that INPP4B also has tumour-promoting properties in cancers including acute myeloid leukemia, colon cancer, melanoma and breast cancer. Together these findings suggest that INPP4B may be a context dependent cancer gene. Whether INPP4B functions solely in a tumour suppressing or tumour promoting manner, or both in non-transformed cells is currently not clear. In this study, consequences of deficiency and overexpression of INPP4B on cellular transformation was investigated using a mouse embryonic fibroblast (MEF) model of cellular transformation. We observed that neither deficiency nor overexpression of INPP4B was sufficient to induce neoplastic transformation, alone or in combination with H-Ras V12 or E1A overexpression. However, Inpp4b-deficiency did cooperate with SV40 T-Large-mediated cellular transformation, a finding which was associated with increased phosphorylated-Akt levels. Transformation and phosphorylated-Akt levels were dampened upon overexpression of INPP4B in SV40 T-Large-MEF. Together, our findings support a model where INPP4B function suppresses transformation mediated by SV40 T-Large, but is inconsequential for Ras and E1A mediated transformation.
Collapse
Affiliation(s)
| | - Keyue Chen
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Brittany Salmon
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal, Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Bellio M, Caux M, Vauclard A, Chicanne G, Gratacap MP, Terrisse AD, Severin S, Payrastre B. Phosphatidylinositol 3 monophosphate metabolizing enzymes in blood platelet production and in thrombosis. Adv Biol Regul 2019; 75:100664. [PMID: 31604685 DOI: 10.1016/j.jbior.2019.100664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/09/2023]
Abstract
Blood platelets, produced by the fragmentation of megakaryocytes, play a key role in hemostasis and thrombosis. Being implicated in atherothrombosis and other thromboembolic disorders, they represent a major therapeutic target for antithrombotic drug development. Several recent studies have highlighted an important role for the lipid phosphatidylinositol 3 monophosphate (PtdIns3P) in megakaryocytes and platelets. PtdIns3P, present in small amounts in mammalian cells, is involved in the control of endocytic trafficking and autophagy. Its metabolism is finely regulated by specific kinases and phosphatases. Class II (α, β and γ) and III (Vps34) phosphoinositide-3-kinases (PI3Ks), INPP4 and Fig4 are involved in the production of PtdIns3P whereas PIKFyve, myotubularins (MTMs) and type II PIPK metabolize PtdIns3P. By regulating the turnover of different pools of PtdIns3P, class II (PI3KC2α) and class III (Vps34) PI3Ks have been recently involved in the regulation of platelet production and functions. These pools of PtdIns3P appear to modulate membrane organization and intracellular trafficking. Moreover, PIKFyve and INPP4 have been recently implicated in arterial thrombosis. In this review, we will discuss the role of PtdIns3P metabolizing enzymes in platelet production and function. Potential new anti-thrombotic therapeutic perspectives based on inhibitors targeting specifically PtdIns3P metabolizing enzymes will also be commented.
Collapse
Affiliation(s)
- Marie Bellio
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Manuella Caux
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Alicia Vauclard
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Gaëtan Chicanne
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Marie-Pierre Gratacap
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Anne-Dominique Terrisse
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Sonia Severin
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Bernard Payrastre
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Laboratoire d'Hématologie, Hopital Universitaire de Toulouse, Toulouse, France.
| |
Collapse
|
18
|
Zhang M, Suarez E, Vasquez JL, Nathanson L, Peterson LE, Rajapakshe K, Basil P, Weigel NL, Coarfa C, Agoulnik IU. Inositol polyphosphate 4-phosphatase type II regulation of androgen receptor activity. Oncogene 2018; 38:1121-1135. [PMID: 30228349 PMCID: PMC6377303 DOI: 10.1038/s41388-018-0498-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/05/2018] [Accepted: 08/24/2018] [Indexed: 11/18/2022]
Abstract
Activation and transcriptional reprogramming of AR in advanced prostate cancer frequently coincides with the loss of two tumor suppressors, INPP4B and PTEN, which are highly expressed in human and mouse prostate epithelium. While regulation of AR signaling by PTEN has been described by multiple groups, it is not known whether the loss of INPP4B affects AR activity. Using prostate cancer cell lines we showed that INPP4B regulates AR transcriptional activity and the oncogenic signaling pathways Akt and PKC. Analysis of gene expression in prostate cancer patient cohorts showed a positive correlation between INPP4B expression and both AR mRNA levels and AR transcriptional output. Using an Inpp4b-/- mouse model, we demonstrated that INPP4B suppresses Akt and PKC signaling pathways and modulates AR transcriptional activity in normal mouse prostate. Remarkably, PTEN protein levels and phosphorylation of S380 were the same in Inpp4b-/- and WT males, suggesting that the observed changes were due exclusively to the loss of INPP4B. Our data show that INPP4B modulates AR activity in normal prostate and its loss contributes to the AR-dependent transcriptional profile in prostate cancer.
Collapse
Affiliation(s)
- Manqi Zhang
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, 33199, USA
| | - Egla Suarez
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Judy L Vasquez
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | | | - Leif E Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Paul Basil
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nancy L Weigel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Irina U Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Biomolecular Science Institute, School of Integrated Science and Humanity, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
19
|
Kang JH, Lim H, Lee DS, Yim M. Montelukast inhibits RANKL‑induced osteoclast formation and bone loss via CysLTR1 and P2Y12. Mol Med Rep 2018; 18:2387-2398. [PMID: 29916540 DOI: 10.3892/mmr.2018.9179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/17/2018] [Indexed: 11/06/2022] Open
Abstract
Osteoclasts (OCs) are resorptive cells responsible for bone erosion in diseases, including osteoporosis, periodontitis and rheumatoid arthritis. Montelukast is a cysteinyl leukotriene receptor 1 (CysLTR1) antagonist clinically used for the treatment of asthma. In the present study, the role of CysLTR1 on OC formation and bone loss was investigated using montelukast. Montelukast inhibited receptor activator of nuclear factor‑κB ligand (RANKL)‑induced OC formation in cultures of mouse bone marrow macrophages. Additionally, montelukast suppressed actin ring formation and bone resorption activity of differentiated OCs. The inhibitory effect of montelukast was associated with impaired activation of extracellular signal‑regulated kinase, AKT serine/threonine kinase, and/or phospholipase Cγ2 signaling pathways downstream of RANK, followed by decreased expression of nuclear factor of activated T cells c1. Notably, OC formation was efficiently restored by addition of adenosine diphosphate, a P2Y12 agonist, as well as by addition of CysLT. Furthermore, similar to montelukast, P2Y12 blockade by a pharmacological inhibitor or siRNAs suppressed OC differentiation. These data indicate the involvement of the P2Y12 receptor in the inhibitory effect of montelukast on osteoclastogenesis. In vivo, montelukast significantly inhibited inflammation‑induced osteoclastogenesis in the calvarial model. Montelukast also served a protective role in a murine ovariectomy (OVX)‑ and unloading‑induced bone loss model. Altogether, these results confirmed that the CysLTR1 antagonist exerted an inhibitory effect on OC formation in vitro and in vivo. It may be useful for the treatment of bone diseases associated with excessive bone resorption.
Collapse
Affiliation(s)
- Ju-Hee Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Sookmyung Women's University, Seoul 140‑742, Republic of Korea
| | - Hyungsik Lim
- Department of Physics, Hunter College of The City University of New York, New York, NY 10065, USA
| | - Dong-Seok Lee
- Department of Natural Sciences, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mijung Yim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Sookmyung Women's University, Seoul 140‑742, Republic of Korea
| |
Collapse
|
20
|
Pata M, Vacher J. Ostm1 Bifunctional Roles in Osteoclast Maturation: Insights From a Mouse Model Mimicking a Human OSTM1 Mutation. J Bone Miner Res 2018; 33:888-898. [PMID: 29297601 DOI: 10.1002/jbmr.3378] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 11/12/2022]
Abstract
Ostm1 mutations are responsible for the most severe form of osteopetrosis in human and mice. To gain insight into Ostm1 cellular functions, we engineered a conditional in-frame deletion of the Ostm1 transmembrane domain and generated the first Ostm1 mouse model with a human mutation. Systemic targeting of Ostm1 loss of transmembrane domain produced osteopetrosis, as in the null Ostm1 gl/gl mouse. Significantly, conditional osteoclast targeting of Ostm1 resulted in similar osteopetrosis, thereby demonstrating that the intrinsic Ostm1 osteoclast deficiency is solely responsible for the mouse phenotype. Our analysis showed oversized osteoclasts with enhanced multinucleation associated with stimulation of intracellular calcium levels, of Nfatc1 nuclear re-localization, and of specific downstream Nfatc1 target genes, providing compelling evidence that Ostm1 is a negative regulator of preosteoclast fusion. Moreover, mature OCs with Ostm1 loss of transmembrane domain show appropriate levels of intracellular acidification but an altered distribution pattern, highlighting misregulation of endolysosome localization and dispersion. Consistently, the hydrolases tartrate-resistant acid phosphatase (TRAP) and cathepsin K (Ctsk) normally produced are sequestered within the osteoclasts and are not extracellularly secreted. These studies defined bifunctional roles for Ostm1 as a major regulator of preosteoclast cytoskeletal rearrangements toward cell multinucleation and of mature osteoclast intracellular lysosomal trafficking and exocytosis mechanism, both of which are essential for bone resorption. Importantly, these Ostm1 molecular and regulatory functions could serve as preclinical targets in this mouse model toward osteoclastogenic pathologies as osteoporosis and inflammation-induced bone loss. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Monica Pata
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| |
Collapse
|
21
|
Abstract
In higher eukaryotes, the Tyr phosphorylation status of cellular proteins results from the coordinated action of Protein Tyrosine Kinases (PTKs) and Protein Tyrosine Phosphatases (PTPs). PTPs have emerged as highly regulated enzymes with diverse substrate specificity, and proteins with Tyr-dephosphorylation or Tyr-dephosphorylation-like properties can be clustered as the PTPome. This includes proteins from the PTP superfamily, which display a Cys-based catalytic mechanism, as well as enzymes from other gene families (Asp-based phosphatases, His-based phosphatases) that have converged in protein Tyr-dephosphorylation-related functions by using non-Cys-based catalytic mechanisms. Within the Cys-based members of the PTPome, classical PTPs dephosphorylate specific phosphoTyr (pTyr) residues from protein substrates, whereas VH1-like dual-specificity PTPs dephosphorylate pTyr, pSer, and pThr residues, as well as nonproteinaceous substrates, including phosphoinositides and phosphorylated carbohydrates. In addition, several PTPs have impaired catalytic activity as a result of amino acid substitutions at their active sites, but retain regulatory functions related with pTyr signaling. As a result of their relevant biological activity, many PTPs are linked to human disease, including cancer, neurodevelopmental, and metabolic diseases, making these proteins important drug targets and molecular markers in the clinic. Here, a brief overview on the biochemistry and physiology of the different groups of proteins that belong to the mammalian PTPome is presented.
Collapse
|
22
|
Macrophages and osteoclasts stem from a bipotent progenitor downstream of a macrophage/osteoclast/dendritic cell progenitor. Blood Adv 2017; 1:1993-2006. [PMID: 29296846 DOI: 10.1182/bloodadvances.2017008540] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Monocytes/macrophages (MΦs), osteoclasts (OCs), and dendritic cells (DCs) are closely related cell types of high clinical significance, but the exact steps in their lineage commitment are unclear. In studies on MΦ and DC development, OC development is generally not addressed. Furthermore, findings on DC development are confusing, because monocytes can also differentiate into DC-like cells. To resolve these issues, we have examined the development of monocytes/MΦs, OCs, and DCs from common progenitors, using the homeostatic driver cytokines macrophage colony-stimulating factor, RANK ligand (L), and Flt3L. In mouse bone marrow, B220-CD11blow/-c-Kit+c-Fms+ cells could be dissected into a CD27+Flt3+ population that proved oligopotent for MΦ/OC/DC development (MODP) and a CD27low/-Flt3- population that proved bipotent for MΦ/OC development (MOP). Developmental potential and relationship of MODP and downstream MOP populations are demonstrated by differentiation cultures, functional analysis of MΦ/OC/DC offspring, and genome-wide messenger RNA expression analysis. A common DC progenitor (CDP) has been described as committed to plasmacytoid and conventional DC development. However, the human CDP proved identical to the MODP population, whereas the mouse CDP largely overlapped with the MODP population and was accordingly oligopotent for MΦ, OC, and DC development. The CX3CR1+ MΦ/DC progenitor (MDP) population described in the mouse generated MΦs and OCs but not DCs. Thus, monocytes/MΦs, OCs, and DCs share a common progenitor that gives rise to a bipotent MΦ/OC progenitor, but a dedicated DC progenitor is currently undefined. The definition of these progenitor populations may serve diagnostics and interventions in diseases with pathogenic activity of MΦs, OCs, or DCs.
Collapse
|
23
|
Zhou R, Lin X, Li DY, Wang XF, Greenbaum J, Chen YC, Zeng CP, Lu JM, Ao ZX, Peng LP, Bai XC, Shen J, Deng HW. Identification of novel genetic loci for osteoporosis and/or rheumatoid arthritis using cFDR approach. PLoS One 2017; 12:e0183842. [PMID: 28854271 PMCID: PMC5576737 DOI: 10.1371/journal.pone.0183842] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/12/2017] [Indexed: 12/19/2022] Open
Abstract
There are co-morbidity between osteoporosis (OP) and rheumatoid arthritis (RA). Some genetic risk factors have been identified for these two phenotypes respectively in previous research; however, they accounted for only a small portion of the underlying total genetic variances. Here, we sought to identify additional common genetic loci associated with OP and/or RA. The conditional false discovery rate (cFDR) approach allows detection of additional genetic factors (those respective ones as well as common pleiotropic ones) for the two associated phenotypes. We collected and analyzed summary statistics provided by large, multi-center GWAS studies of FNK (femoral neck) BMD (a major risk factor for osteoporosis) (n = 53,236) and RA (n = 80,799). The conditional quantile-quantile (Q-Q) plots can assess the enrichment of SNPs related to FNK BMD and RA, respectively. Furthermore, we identified shared loci between FNK BMD and RA using conjunction cFDR (ccFDR). We found strong enrichment of p-values in FNK BMD when conditional Q-Q was done on RA and vice versa. We identified 30 novel OP-RA associated pleiotropic loci that have not been reported in previous OP or RA GWAS, 18 of which located in the MHC (major histocompatibility complex) region previously reported to play an important role in immune system and bone health. We identified some specific novel polygenic factors for OP and RA respectively, and identified 30 novel OP-RA associated pleiotropic loci. These discovery findings may offer novel pathobiological insights, and suggest new targets and pathways for drug development in OP and RA patients.
Collapse
Affiliation(s)
- Rou Zhou
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Ding-You Li
- Department of Gastroenterology, Children’s Mercy Kansas City, University of Missouri Kansas City School of Medicine, Kansas City, MO, United States of America
| | - Xia-Fang Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Jonathan Greenbaum
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, United States of America
| | - Yuan-Cheng Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Chun-Ping Zeng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Jun-Min Lu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Zeng-Xing Ao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Lin-Ping Peng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Xiao Chun Bai
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Hong-Wen Deng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, United States of America
| |
Collapse
|
24
|
Regulation of PI3K effector signalling in cancer by the phosphoinositide phosphatases. Biosci Rep 2017; 37:BSR20160432. [PMID: 28082369 PMCID: PMC5301276 DOI: 10.1042/bsr20160432] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/24/2022] Open
Abstract
Class I phosphoinositide 3-kinase (PI3K) generates phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) at the plasma membrane in response to growth factors, activating a signalling cascade that regulates many cellular functions including cell growth, proliferation, survival, migration and metabolism. The PI3K pathway is commonly dysregulated in human cancer, and drives tumorigenesis by promoting aberrant cell growth and transformation. PtdIns(3,4,5)P3 facilitates the activation of many pleckstrin homology (PH) domain-containing proteins including the serine/threonine kinase AKT. There are three AKT isoforms that are frequently hyperactivated in cancer through mutation, amplification or dysregulation of upstream regulatory proteins. AKT isoforms have converging and opposing functions in tumorigenesis. PtdIns(3,4,5)P3 signalling is degraded and terminated by phosphoinositide phosphatases such as phosphatase and tensin homologue (PTEN), proline-rich inositol polyphosphate 5-phosphatase (PIPP) (INPP5J) and inositol polyphosphate 4-phosphatase type II (INPP4B). PtdIns(3,4,5)P3 is rapidly hydrolysed by PIPP to generate phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2), which is further hydrolysed by INPP4B to form phosphatidylinositol 3-phosphate (PtdIns3P). PtdIns(3,4)P2 and PtdIns3P are also important signalling molecules; PtdIns(3,4)P2 together with PtdIns(3,4,5)P3 are required for maximal AKT activation and PtdIns3P activates PI3K-dependent serum and glucocorticoid-regulated kinase (SGK3) signalling. Loss of Pten, Pipp or Inpp4b expression or function promotes tumour growth in murine cancer models through enhanced AKT isoform-specific signalling. INPP4B inhibits PtdIns(3,4)P2-mediated AKT activation in breast and prostate cancer; however, INPP4B expression is increased in acute myeloid leukaemia (AML), melanoma and colon cancer where it paradoxically promotes cell proliferation, transformation and/or drug resistance. This review will discuss how PTEN, PIPP and INPP4B distinctly regulate PtdIns(3,4,5)P3 signalling downstream of PI3K and how dysregulation of these phosphatases affects cancer outcomes.
Collapse
|
25
|
Bongiorni S, Gruber CEM, Bueno S, Chillemi G, Ferrè F, Failla S, Moioli B, Valentini A. Transcriptomic investigation of meat tenderness in two Italian cattle breeds. Anim Genet 2016; 47:273-87. [DOI: 10.1111/age.12418] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2015] [Indexed: 12/23/2022]
Affiliation(s)
- S. Bongiorni
- Department for Innovation in Biological, Agro-food and Forest systems, DIBAF; University of Tuscia; via S. Camillo de Lelliss.n.c. Viterbo 01100 Italy
| | - C. E. M. Gruber
- Molecular Digital Diagnostics (MDD); s.r.l., via San Camillo de Lellis Viterbo 01100 Italy
| | - S. Bueno
- Cineca Supercomputing Center; via dei Tizii 6 Rome 00185 Italy
| | - G. Chillemi
- Cineca Supercomputing Center; via dei Tizii 6 Rome 00185 Italy
| | - F. Ferrè
- Cineca Supercomputing Center; via dei Tizii 6 Rome 00185 Italy
- Centre for Molecular Bioinformatics; Biology Department; University of Rome Tor Vergata; Rome Italy
| | - S. Failla
- Consiglio per la Ricerca e la sperimentazione in Agricoltura; Monterotondo Italy
| | - B. Moioli
- Consiglio per la Ricerca e la sperimentazione in Agricoltura; Monterotondo Italy
| | - A. Valentini
- Department for Innovation in Biological, Agro-food and Forest systems, DIBAF; University of Tuscia; via S. Camillo de Lelliss.n.c. Viterbo 01100 Italy
| |
Collapse
|
26
|
Alonso A, Pulido R. The extended human PTPome: a growing tyrosine phosphatase family. FEBS J 2015; 283:1404-29. [PMID: 26573778 DOI: 10.1111/febs.13600] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/02/2015] [Accepted: 11/13/2015] [Indexed: 12/13/2022]
Abstract
Tyr phosphatases are, by definition, enzymes that dephosphorylate phospho-Tyr (pTyr) from proteins. This activity is found in several structurally diverse protein families, including the protein Tyr phosphatase (PTP), arsenate reductase, rhodanese, haloacid dehalogenase (HAD) and His phosphatase (HP) families. Most of these families include members with substrate specificity for non-pTyr substrates, such as phospho-Ser/phospho-Thr, phosphoinositides, phosphorylated carbohydrates, mRNAs, or inorganic moieties. A Cys is essential for catalysis in PTPs, rhodanese and arsenate reductase enzymes, whereas this work is performed by an Asp in HAD phosphatases and by a His in HPs, via a catalytic mechanism shared by all of the different families. The category that contains most Tyr phosphatases is the PTP family, which, although it received its name from this activity, includes Ser, Thr, inositide, carbohydrate and RNA phosphatases, as well as some inactive pseudophosphatase proteins. Here, we propose an extended collection of human Tyr phosphatases, which we call the extended human PTPome. The addition of new members (SACs, paladin, INPP4s, TMEM55s, SSU72, and acid phosphatases) to the currently categorized PTP group of enzymes means that the extended human PTPome contains up to 125 proteins, of which ~ 40 are selective for pTyr. We set criteria to ascribe proteins to the extended PTPome, and summarize the more important features of the new PTPome members in the context of their phosphatase activity and their relationship with human disease.
Collapse
Affiliation(s)
- Andrés Alonso
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Rafael Pulido
- Biocruces Health Research Institute, Barakaldo, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
27
|
Mapping of Craniofacial Traits in Outbred Mice Identifies Major Developmental Genes Involved in Shape Determination. PLoS Genet 2015; 11:e1005607. [PMID: 26523602 PMCID: PMC4629907 DOI: 10.1371/journal.pgen.1005607] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/24/2015] [Indexed: 02/05/2023] Open
Abstract
The vertebrate cranium is a prime example of the high evolvability of complex traits. While evidence of genes and developmental pathways underlying craniofacial shape determination is accumulating, we are still far from understanding how such variation at the genetic level is translated into craniofacial shape variation. Here we used 3D geometric morphometrics to map genes involved in shape determination in a population of outbred mice (Carworth Farms White, or CFW). We defined shape traits via principal component analysis of 3D skull and mandible measurements. We mapped genetic loci associated with shape traits at ~80,000 candidate single nucleotide polymorphisms in ~700 male mice. We found that craniofacial shape and size are highly heritable, polygenic traits. Despite the polygenic nature of the traits, we identified 17 loci that explain variation in skull shape, and 8 loci associated with variation in mandible shape. Together, the associated variants account for 11.4% of skull and 4.4% of mandible shape variation, however, the total additive genetic variance associated with phenotypic variation was estimated in ~45%. Candidate genes within the associated loci have known roles in craniofacial development; this includes 6 transcription factors and several regulators of bone developmental pathways. One gene, Mn1, has an unusually large effect on shape variation in our study. A knockout of this gene was previously shown to affect negatively the development of membranous bones of the cranial skeleton, and evolutionary analysis shows that the gene has arisen at the base of the bony vertebrates (Eutelostomi), where the ossified head first appeared. Therefore, Mn1 emerges as a key gene for both skull formation and within-population shape variation. Our study shows that it is possible to identify important developmental genes through genome-wide mapping of high-dimensional shape features in an outbred population. Formation of the face, mandible, and skull is determined in part by genetic factors, but the relationship between genetic variation and craniofacial development is not well understood. We demonstrate how recent advances in mouse genomics and statistical methods can be used to identify genes involved in craniofacial development. We use outbred mice together with a dense panel of genetic markers to identify genetic loci affecting craniofacial shape. Some of the loci we identify are also known from past studies to contribute to craniofacial development and bone formation. For example, the top candidate gene identified in this study, Mn1, is a gene that appeared at a time when animals started to form bony skulls, suggesting that it may be a key gene in this evolutionary innovation. This further suggests that Mn1 and other genes involved in head formation are also responsible for more fine-grained regulation of its shape. Our results confirm that the outbred mouse population used in this study is suitable to identify single genetic factors even under conditions where many genes cooperate to generate a complex phenotype.
Collapse
|
28
|
Phosphatidylinositol (3,4) bisphosphate-specific phosphatases and effector proteins: A distinct branch of PI3K signaling. Cell Signal 2015; 27:1789-98. [DOI: 10.1016/j.cellsig.2015.05.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/16/2015] [Accepted: 05/20/2015] [Indexed: 01/22/2023]
|
29
|
Hodgson MC, Deryugina EI, Suarez E, Lopez SM, Lin D, Xue H, Gorlov IP, Wang Y, Agoulnik IU. INPP4B suppresses prostate cancer cell invasion. Cell Commun Signal 2014. [PMID: 25248616 DOI: 10.1186/preaccept-2663637391256502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND INPP4B and PTEN dual specificity phosphatases are frequently lost during progression of prostate cancer to metastatic disease. We and others have previously shown that loss of INPP4B expression correlates with poor prognosis in multiple malignancies and with metastatic spread in prostate cancer. RESULTS We demonstrate that de novo expression of INPP4B in highly invasive human prostate carcinoma PC-3 cells suppresses their invasion both in vitro and in vivo. Using global gene expression analysis, we found that INPP4B regulates a number of genes associated with cell adhesion, the extracellular matrix, and the cytoskeleton. Importantly, de novo expressed INPP4B suppressed the proinflammatory chemokine IL-8 and induced PAK6. These genes were regulated in a reciprocal manner following downregulation of INPP4B in the independently derived INPP4B-positive LNCaP prostate cancer cell line. Inhibition of PI3K/Akt pathway, which is highly active in both PC-3 and LNCaP cells, did not reproduce INPP4B mediated suppression of IL-8 mRNA expression in either cell type. In contrast, inhibition of PKC signaling phenocopied INPP4B-mediated inhibitory effect on IL-8 in either prostate cancer cell line. In PC-3 cells, INPP4B overexpression caused a decline in the level of metastases associated BIRC5 protein, phosphorylation of PKC, and expression of the common PKC and IL-8 downstream target, COX-2. Reciprocally, COX-2 expression was increased in LNCaP cells following depletion of endogenous INPP4B. CONCLUSION Taken together, we discovered that INPP4B is a novel suppressor of oncogenic PKC signaling, further emphasizing the role of INPP4B in maintaining normal physiology of the prostate epithelium and suppressing metastatic potential of prostate tumors.
Collapse
|
30
|
Abstract
Background INPP4B and PTEN dual specificity phosphatases are frequently lost during progression of prostate cancer to metastatic disease. We and others have previously shown that loss of INPP4B expression correlates with poor prognosis in multiple malignancies and with metastatic spread in prostate cancer. Results We demonstrate that de novo expression of INPP4B in highly invasive human prostate carcinoma PC-3 cells suppresses their invasion both in vitro and in vivo. Using global gene expression analysis, we found that INPP4B regulates a number of genes associated with cell adhesion, the extracellular matrix, and the cytoskeleton. Importantly, de novo expressed INPP4B suppressed the proinflammatory chemokine IL-8 and induced PAK6. These genes were regulated in a reciprocal manner following downregulation of INPP4B in the independently derived INPP4B-positive LNCaP prostate cancer cell line. Inhibition of PI3K/Akt pathway, which is highly active in both PC-3 and LNCaP cells, did not reproduce INPP4B mediated suppression of IL-8 mRNA expression in either cell type. In contrast, inhibition of PKC signaling phenocopied INPP4B-mediated inhibitory effect on IL-8 in either prostate cancer cell line. In PC-3 cells, INPP4B overexpression caused a decline in the level of metastases associated BIRC5 protein, phosphorylation of PKC, and expression of the common PKC and IL-8 downstream target, COX-2. Reciprocally, COX-2 expression was increased in LNCaP cells following depletion of endogenous INPP4B. Conclusion Taken together, we discovered that INPP4B is a novel suppressor of oncogenic PKC signaling, further emphasizing the role of INPP4B in maintaining normal physiology of the prostate epithelium and suppressing metastatic potential of prostate tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12964-014-0061-y) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
Abstract
The specific interaction of phosphoinositides with proteins is critical for a plethora of cellular processes, including cytoskeleton remodelling, mitogenic signalling, ion channel regulation and membrane traffic. The spatiotemporal restriction of different phosphoinositide species helps to define compartments within the cell, and this is particularly important for membrane trafficking within both the secretory and endocytic pathways. Phosphoinositide homoeostasis is tightly regulated by a large number of inositol kinases and phosphatases, which respectively phosphorylate and dephosphorylate distinct phosphoinositide species. Many of these enzymes have been implicated in regulating membrane trafficking and, accordingly, their dysregulation has been linked to a number of human diseases. In the present review, we focus on the inositol phosphatases, concentrating on their roles in membrane trafficking and the human diseases with which they have been associated.
Collapse
|
32
|
Ding H, Sun Y, Hou Y, Li L. Effects of INPP4B gene transfection combined with PARP inhibitor on castration therapy—Resistant prostate cancer cell line, PC3. Urol Oncol 2014; 32:720-6. [DOI: 10.1016/j.urolonc.2013.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/26/2013] [Accepted: 11/26/2013] [Indexed: 02/03/2023]
|
33
|
Zheng T, Wang X, Yim M. Miconazole inhibits receptor activator of nuclear factor-κB ligand-mediated osteoclast formation and function. Eur J Pharmacol 2014; 737:185-93. [PMID: 24842191 DOI: 10.1016/j.ejphar.2014.04.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/18/2022]
Abstract
Osteoclasts are responsible for bone erosion in diseases as diverse as osteoporosis, periodontitis, and rheumatoid arthritis. Antifungal products have received recent attention as potential therapeutic and preventative drugs in human disease. Since little is known about the action of miconazole, an antifungal imidazole, on bone metabolism, we investigated the effects of miconazole on osteoclast formation using mouse bone marrow macrophages (BMMs). Miconazole inhibited RANKL-induced osteoclast formation in a dose-dependent manner without cytotoxicity. Furthermore, miconazole inhibited the bone resorptive activity of osteoclasts. Miconazole suppressed RANKL-induced expression of c-Fos and NFATc1, two essential transcription factors for osteoclast differentiation. Miconazole seemed to inhibit osteoclast formation MAPK pathways as well as Blimp1 through MafB expression. Miconazole also inhibited RANKL-induced expression of the pro-inflammatory cytokines, COX-2 and iNOS. In accordance with the in vitro study, miconazole reduced lipopolysaccharide-induced osteoclast formation in vivo. Therefore, miconazole exerted an inhibitory effect on osteoclast formation in vitro and in vivo. It could be useful for the treatment of bone diseases associated with excessive bone resorption.
Collapse
Affiliation(s)
- Ting Zheng
- College of Pharmacy, Sookmyung Women׳s University, Hyochangwongil 52, Yongsan-ku, Seoul 140-742, Republic of Korea
| | - Xin Wang
- College of Pharmacy, Sookmyung Women׳s University, Hyochangwongil 52, Yongsan-ku, Seoul 140-742, Republic of Korea
| | - Mijung Yim
- College of Pharmacy, Sookmyung Women׳s University, Hyochangwongil 52, Yongsan-ku, Seoul 140-742, Republic of Korea.
| |
Collapse
|
34
|
Zhang X, Chen H, Wang X, Zhao W, Chen JJ. Expression and transcriptional profiling of the LKB1 tumor suppressor in cervical cancer cells. Gynecol Oncol 2014; 134:372-8. [PMID: 24792998 DOI: 10.1016/j.ygyno.2014.04.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To characterize the biological activities of LKB1, examine LKB1 protein expression and identify LKB1-regulated genes that may serve as therapeutic targets in cervical cancer. METHODS Proliferation of cervical cancer HeLa cells expressing LKB1 was examined. LKB1 expression in normal cervical tissues and cervical cancers was assessed by immunohistochemistry. Gene expression profiles of cervical cancer HeLa cells stably expressing LKB1 were analyzed by microarray. Differentially expressed genes were analyzed using Gene Ontology (GO) terms and the Kyoto Encyclopedia of Genes and Genomes (KEGG) PATHWAY database. Quantitative RT-PCR was used to validate the microarray data. The expression of lipid phosphatase inositol polyphosphate 4-phosphatase type II (INPP4B) was confirmed by western blotting. RESULTS Expression of LKB1 inhibited HeLa cell proliferation, activated AMPK and was lost in more than 50% of cervical carcinomas. More than 200 genes were differentially expressed between HeLa cells with and without LKB1. Bioinformatics analysis with GO annotation indicated that LKB1 plays a role in receiving diverse stimuli and converting them into molecular signals. KEGG PATHWAY analysis showed that 8 pathways were significantly regulated. These include arginine and proline metabolism and inositol phosphate metabolism. The differential expression of 7 randomly selected genes was confirmed by quantitative RT-PCR. Furthermore, the steady-state level of INPP4B protein was up-regulated in LKB1-overexpressing cells. CONCLUSIONS This study establishes LKB1 as an important tumor suppressor in cervical cancer and sheds light on a novel signaling pathway regulated by LKB1.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Microbiology, Shandong University School of Medicine, Jinan, Shandong, China; Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01532, USA
| | - Hanxiang Chen
- Department of Microbiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiao Wang
- Department of Pathology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Weiming Zhao
- Department of Microbiology, Shandong University School of Medicine, Jinan, Shandong, China.
| | - Jason J Chen
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01532, USA; Cancer Research Center, Shandong University School of Medicine, Jinan, Shandong, China.
| |
Collapse
|
35
|
Sun Y, Ding H, Liu X, Li X, Li L. INPP4B overexpression enhances the antitumor efficacy of PARP inhibitor AG014699 in MDA-MB-231 triple-negative breast cancer cells. Tumour Biol 2014; 35:4469-77. [DOI: 10.1007/s13277-013-1589-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022] Open
|
36
|
Bertucci MC, Mitchell CA. Phosphoinositide 3-kinase and INPP4B in human breast cancer. Ann N Y Acad Sci 2013; 1280:1-5. [PMID: 23551093 DOI: 10.1111/nyas.12036] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The PI3K/Akt signaling pathway is frequently increased in many human cancers, including breast cancer. Recent studies have identified INPP4B, which inhibits PI3K signaling, as an emerging tumor suppressor in breast cancer. This short review discusses these issues and the possibility that INPP4B is an important regulator in many cancers.
Collapse
Affiliation(s)
- Micka C Bertucci
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
37
|
Vacher J. Inpp4b is a novel negative modulator of osteoclast differentiation and a prognostic locus for human osteoporosis. Ann N Y Acad Sci 2013; 1280:52-4. [PMID: 23551105 DOI: 10.1111/nyas.12014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inositol polyphosphate 4-phosphatase type II (Inpp4b) is a novel negative modulator of osteoclast differentiation and a prognostic locus for human osteoporosis. This short overview summarizes some of the cellular, molecular, and crosstalk signaling mechanisms that control osteoclast and osteoblast differentiation and activation.
Collapse
Affiliation(s)
- Jean Vacher
- Clinical Research Institute of Montreal, Départment de Médecine, Université de Montréal, Department of Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
38
|
Lopez SM, Hodgson MC, Packianathan C, Bingol-Ozakpinar O, Uras F, Rosen BP, Agoulnik IU. Determinants of the tumor suppressor INPP4B protein and lipid phosphatase activities. Biochem Biophys Res Commun 2013; 440:277-282. [PMID: 24070612 PMCID: PMC4578723 DOI: 10.1016/j.bbrc.2013.09.077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 02/08/2023]
Abstract
The tumor suppressor INPP4B is an important regulator of phosphatidyl-inositol signaling in the cell. Reduced INPP4B expression is associated with poor outcomes for breast, prostate, and ovarian cancer patients. INPP4B contains a CX5R catalytic motif characteristic of dual-specificity phosphatases, such as PTEN. Lipid phosphatase activity of INPP4B has previously been described. In this report we show that INPP4B can dephosphorylate para-nitrophenyl phosphate (pNPP) and 6,8-difluoro-4-methylumbelliferyl (DiFMUP), synthetic phosphotyrosine analogs, suggesting that INPP4B has protein tyrosine phosphatase (PTP) activity. Using mutagenesis, we examined the functional role of specific amino acids within the INPP4B C842KSAKDR catalytic site. The K843M mutant displayed increased pNPP hydrolysis, the K846M mutant lost lipid phosphatase activity with no effect on PTP activity, and the D847E substitution ablated PTP activity and significantly reduced lipid phosphatase activity. Further, we show that INPP4B but not PTEN is able to reduce tyrosine phosphorylation of Akt1 and both the lipid and PTP activity of INPP4B likely contribute to the reduction of Akt1 phosphorylation. Taken together our data identified key residues in the INPP4B catalytic domain associated with lipid and protein phosphatase activities and found a robust downstream target regulated by INPP4B but not PTEN.
Collapse
Affiliation(s)
- Sandra M. Lopez
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
| | - Myles C. Hodgson
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
| | - Charles Packianathan
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
| | - Ozlem Bingol-Ozakpinar
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
- Department of Biochemistry, Marmara University School of Pharmacy, Haydarpasa, 34668 Istanbul, Turkey
| | - Fikriye Uras
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
- Department of Biochemistry, Marmara University School of Pharmacy, Haydarpasa, 34668 Istanbul, Turkey
| | - Barry P. Rosen
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
| | - Irina U. Agoulnik
- Department of Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St. Miami, FL 33199, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
39
|
Ferron M, Settembre C, Shimazu J, Lacombe J, Kato S, Rawlings DJ, Ballabio A, Karsenty G. A RANKL-PKCβ-TFEB signaling cascade is necessary for lysosomal biogenesis in osteoclasts. Genes Dev 2013; 27:955-69. [PMID: 23599343 DOI: 10.1101/gad.213827.113] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bone resorption by osteoclasts requires a large number of lysosomes that release proteases in the resorption lacuna. Whether lysosomal biogenesis is a consequence of the action of transcriptional regulators of osteoclast differentiation or is under the control of a different and specific transcriptional pathway remains unknown. We show here, through cell-based assays and cell-specific gene deletion experiments in mice, that the osteoclast differentiation factor RANKL promotes lysosomal biogenesis once osteoclasts are differentiated through the selective activation of TFEB, a member of the MITF/TFE family of transcription factors. This occurs following PKCβ phosphorylation of TFEB on three serine residues located in its last 15 amino acids. This post-translational modification stabilizes and increases the activity of this transcription factor. Supporting these biochemical observations, mice lacking in osteoclasts--either TFEB or PKCβ--show decreased lysosomal gene expression and increased bone mass. Altogether, these results uncover a RANKL-dependent signaling pathway taking place in differentiated osteoclasts and culminating in the activation of TFEB to enhance lysosomal biogenesis-a necessary step for proper bone resorption.
Collapse
Affiliation(s)
- Mathieu Ferron
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Phosphoinositide signalling molecules interact with a plethora of effector proteins to regulate cell proliferation and survival, vesicular trafficking, metabolism, actin dynamics and many other cellular functions. The generation of specific phosphoinositide species is achieved by the activity of phosphoinositide kinases and phosphatases, which phosphorylate and dephosphorylate, respectively, the inositol headgroup of phosphoinositide molecules. The phosphoinositide phosphatases can be classified as 3-, 4- and 5-phosphatases based on their specificity for dephosphorylating phosphates from specific positions on the inositol head group. The SAC phosphatases show less specificity for the position of the phosphate on the inositol ring. The phosphoinositide phosphatases regulate PI3K/Akt signalling, insulin signalling, endocytosis, vesicle trafficking, cell migration, proliferation and apoptosis. Mouse knockout models of several of the phosphoinositide phosphatases have revealed significant physiological roles for these enzymes, including the regulation of embryonic development, fertility, neurological function, the immune system and insulin sensitivity. Importantly, several phosphoinositide phosphatases have been directly associated with a range of human diseases. Genetic mutations in the 5-phosphatase INPP5E are causative of the ciliopathy syndromes Joubert and MORM, and mutations in the 5-phosphatase OCRL result in Lowe's syndrome and Dent 2 disease. Additionally, polymorphisms in the 5-phosphatase SHIP2 confer diabetes susceptibility in specific populations, whereas reduced protein expression of SHIP1 is reported in several human leukaemias. The 4-phosphatase, INPP4B, has recently been identified as a tumour suppressor in human breast and prostate cancer. Mutations in one SAC phosphatase, SAC3/FIG4, results in the degenerative neuropathy, Charcot-Marie-Tooth disease. Indeed, an understanding of the precise functions of phosphoinositide phosphatases is not only important in the context of normal human physiology, but to reveal the mechanisms by which these enzyme families are implicated in an increasing repertoire of human diseases.
Collapse
|
41
|
Macrophage fusion is controlled by the cytoplasmic protein tyrosine phosphatase PTP-PEST/PTPN12. Mol Cell Biol 2013; 33:2458-69. [PMID: 23589331 DOI: 10.1128/mcb.00197-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Macrophages can undergo cell-cell fusion, leading to the formation of multinucleated giant cells and osteoclasts. This process is believed to promote the proteolytic activity of macrophages toward pathogens, foreign bodies, and extracellular matrices. Here, we examined the role of PTP-PEST (PTPN12), a cytoplasmic protein tyrosine phosphatase, in macrophage fusion. Using a macrophage-targeted PTP-PEST-deficient mouse, we determined that PTP-PEST was not needed for macrophage differentiation or cytokine production. However, it was necessary for interleukin-4-induced macrophage fusion into multinucleated giant cells in vitro. It was also needed for macrophage fusion following implantation of a foreign body in vivo. Moreover, in the RAW264.7 macrophage cell line, PTP-PEST was required for receptor activator of nuclear factor kappa-B ligand (RANKL)-triggered macrophage fusion into osteoclasts. PTP-PEST had no impact on expression of fusion mediators such as β-integrins, E-cadherin, and CD47, which enable macrophages to become fusion competent. However, it was needed for polarization of macrophages, migration induced by the chemokine CC chemokine ligand 2 (CCL2), and integrin-induced spreading, three key events in the fusion process. PTP-PEST deficiency resulted in specific hyperphosphorylation of the protein tyrosine kinase Pyk2 and the adaptor paxillin. Moreover, a fusion defect was induced upon treatment of normal macrophages with a Pyk2 inhibitor. Together, these data argue that macrophage fusion is critically dependent on PTP-PEST. This function is seemingly due to the ability of PTP-PEST to control phosphorylation of Pyk2 and paxillin, thereby regulating cell polarization, migration, and spreading.
Collapse
|
42
|
Gumbleton M, Kerr WG. Role of inositol phospholipid signaling in natural killer cell biology. Front Immunol 2013; 4:47. [PMID: 23508471 PMCID: PMC3589743 DOI: 10.3389/fimmu.2013.00047] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/08/2013] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells are important for host defense against malignancy and infection. At a cellular level NK cells are activated when signals from activating receptors exceed signaling from inhibitory receptors. At a molecular level NK cells undergo an education process to both prevent autoimmunity and acquire lytic capacity. Mouse models have shown important roles for inositol phospholipid signaling in lymphocytes. NK cells from mice with deletion in different members of the inositol phospholipid signaling pathway exhibit defects in development, NK cell repertoire expression and effector function. Here we review the current state of knowledge concerning the function of inositol phospholipid signaling components in NK cell biology.
Collapse
Affiliation(s)
- Matthew Gumbleton
- Department of Microbiology and Immunology, State University of New York Upstate Medical University Syracuse, NY, USA
| | | |
Collapse
|
43
|
Tmem64 modulates calcium signaling during RANKL-mediated osteoclast differentiation. Cell Metab 2013; 17:249-60. [PMID: 23395171 PMCID: PMC3569742 DOI: 10.1016/j.cmet.2013.01.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 12/03/2012] [Accepted: 01/08/2013] [Indexed: 12/14/2022]
Abstract
Osteoclast maturation and function primarily depend on receptor activator of NF-κB ligand (RANKL)-mediated induction of nuclear factor of activated T cells c1 (NFATc1), which is further activated via increased intracellular calcium ([Ca(2+)](i)) oscillation. However, the coordination mechanism that mediates Ca(2+) oscillation during osteoclastogenesis remains ill defined. Here, we identified transmembrane protein 64 (Tmem64) as a regulator of Ca(2+) oscillation during osteoclastogenesis. We found that Tmem64-deficient mice exhibit increased bone mass due in part to impaired osteoclast formation. Using in vitro osteoclast culture systems, we show here that Tmem64 interacts with sarcoplasmic endoplasmic reticulum Ca(2+) ATPase 2 (SERCA2) and modulates its activity. Consequently, Tmem64 deficiency significantly diminishes RANKL-induced [Ca(2+)](i) oscillation, which results in reduced Ca(2+)/calmodulin-dependent protein kinases (CaMK) IV and mitochondrial ROS, both of which contribute to achieving the CREB activity necessary for osteoclast formation. These data demonstrate that Tmem64 is a positive modulator of osteoclast differentiation via SERCA2-dependent Ca(2+) signaling.
Collapse
|
44
|
Oikawa T, Kuroda Y, Matsuo K. Regulation of osteoclasts by membrane-derived lipid mediators. Cell Mol Life Sci 2013; 70:3341-53. [PMID: 23296124 PMCID: PMC3753467 DOI: 10.1007/s00018-012-1238-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/05/2012] [Accepted: 12/10/2012] [Indexed: 12/22/2022]
Abstract
Osteoclasts are bone-resorbing cells of monocytic origin. An imbalance between bone formation and resorption can lead to osteoporosis or osteopetrosis. Osteoclastogenesis is triggered by RANKL- and IP3-induced Ca2+ influx followed by activation of NFATc1, a master transcription factor for osteoclastogenic gene regulation. During differentiation, osteoclasts undergo cytoskeletal remodeling to migrate and attach to the bone surface. Simultaneously, they fuse with each other to form multinucleated cells. These processes require PI3-kinase-dependent cytoskeletal protein activation to initiate cytoskeletal remodeling, resulting in the formation of circumferential podosomes and fusion-competent protrusions. In multinucleated osteoclasts, circumferential podosomes mature into stabilized actin rings, which enables the formation of a ruffled border where intensive membrane trafficking is executed. Membrane lipids, especially phosphoinositides, are key signaling molecules that regulate osteoclast morphology and act as second messengers and docking sites for multiple important effectors. We examine the critical roles of phosphoinositides in the signaling cascades that regulate osteoclast functions.
Collapse
Affiliation(s)
- Tsukasa Oikawa
- Laboratory of Cell and Tissue Biology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | | | | |
Collapse
|
45
|
Balakrishnan A, Chaillet JR. Role of the inositol polyphosphate-4-phosphatase type II Inpp4b in the generation of ovarian teratomas. Dev Biol 2012; 373:118-29. [PMID: 23078915 DOI: 10.1016/j.ydbio.2012.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/14/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
Abstract
Teratomas are a unique class of tumors composed of ecto-, meso- and endodermal tissues, all foreign to the site of origin. In humans, the most common teratoma is the ovarian teratoma. Not much is known about the molecular and genetic etiologies of these tumors. Female carriers of the Tgkd transgene are highly susceptible to developing teratomas. Ovaries of Tgkd/+ hemizygous female mice exhibit defects in luteinization, with numerous corpora lutea, some of which contain central trapped, fully-grown oocytes. Genetically, Tgkd teratomas originate from mature oocytes that have completed meiosis I, suggesting that Tgkd teratomas originate from these trapped oocytes. The insertion of Tgkd 3' of the Inpp4b gene is associated with decreased expression of Inpp4b and changes in intracellular PI3 Kinase/AKT signaling in follicular granulosa cells. Because Inpp4b is not expressed in fully-grown wild-type or Tgkd oocytes, these findings suggest that hyperactivation of the PI3K/AKT pathway caused by the decrease in INPP4B in granulosa cells promotes an ovarian environment defective in folliculogenesis and conducive to teratoma formation.
Collapse
Affiliation(s)
- Ashwini Balakrishnan
- Department of Microbiology and Molecular Genetics, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15208, USA
| | | |
Collapse
|
46
|
Vacher J. [Inositol phosphatase and bone mass: role of INPP4b]. Med Sci (Paris) 2012; 28:152-3. [PMID: 22377302 DOI: 10.1051/medsci/2012282012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|