1
|
Nishida M, Sato A, Shimizu A, Rahman N, Wada A, Kageyama S, Ogita H. EphA-Mediated Regulation of Stomatin Expression in Prostate Cancer Cells. Cancer Med 2024; 13:e70276. [PMID: 39377541 PMCID: PMC11459579 DOI: 10.1002/cam4.70276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND AND AIMS Tumor growth and progression are affected by interactions between tumor cells and stromal cells within the tumor microenvironment. We previously showed that the expression of an integral membrane protein, called stomatin, was increased in cancer cells following their association with stromal cells. Additionally, stomatin impaired the Akt signaling pathway to suppress tumor growth. However, it remains unclear how stomatin expression is regulated. To explore this, we examined the cell surface molecules that can transduce the intercellular communication signals between cancer cells and stromal cells. RESULTS Among these molecules, EphA3 and EphA7 receptors and their ligand ephrin-A5 were found to be expressed in prostate cancer cells, but not in prostate stromal cells. Cell-to-cell contact of prostate cancer cells through the EphA-ephrin-A interaction suppressed stomatin expression, while knockdown of EphA3/7 or ephrin-A5 increased stomatin expression. This increase contributed to an inhibition of prostate cancer cell proliferation. Intracellularly, the binding of ephrin-A to EphA attenuated extracellular signaling-regulated kinase (ERK) activation that promoted stomatin expression. Furthermore, ELK1 and ELK4, which are Ets family transcription factors phosphorylated by ERK, were involved in the induction of stomatin expression. We also found that higher Gleason score prostate cancer tissue samples had increased activation of EphA, while the stomatin expression and activated ERK and ELK levels were all low. In the mouse xenograft tumor samples generated by implantation of prostate cancer cells, EphA3 phosphorylation was attenuated and the ERK-ELK signaling and stomatin expression were enhanced in the area where stromal cells infiltrated the tumor. CONCLUSION The EphA-mediated signaling suppresses the ERK-ELK pathway, leading to the reduction of stomatin expression that affects prostate cancer malignancy.
Collapse
Affiliation(s)
- Masanari Nishida
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Nor Idayu A. Rahman
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akinori Wada
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Susumu Kageyama
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
2
|
Chen SY, Cao JL, Li KP, Wan S, Yang L. BIN1 in cancer: biomarker and therapeutic target. J Cancer Res Clin Oncol 2023; 149:7933-7944. [PMID: 36890396 DOI: 10.1007/s00432-023-04673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/28/2023] [Indexed: 03/10/2023]
Abstract
BACKGROUND The bridging integrator 1 (BIN1) protein was originally identified as a pro-apoptotic tumor suppressor that binds to and inhibits oncogenic MYC transcription factors. BIN1 has complex physiological functions participating in endocytosis, membrane cycling, cytoskeletal regulation, DNA repair deficiency, cell-cycle arrest, and apoptosis. The expression of BIN1 is closely related to the development of various diseases such as cancer, Alzheimer's disease, myopathy, heart failure, and inflammation. PURPOSE Because BIN1 is commonly expressed in terminally differentiated normal tissues and is usually undetectable in refractory or metastatic cancer tissues, this differential expression has led us to focus on human cancers associated with BIN1. In this review, we discuss the potential pathological mechanisms of BIN1 during cancer development and its feasibility as a prognostic marker and therapeutic target for related diseases based on recent findings on its molecular, cellular, and physiological roles. CONCLUSION BIN1 is a tumor suppressor that regulates cancer development through a series of signals in tumor progression and microenvironment. It also makes BIN1 a feasible early diagnostic or prognostic marker for cancer.
Collapse
Affiliation(s)
- Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jin-Long Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Shun Wan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
3
|
Thomas S, Dilbarov N, Kelly J, Mercogliano G, Prendergast GC. Diet effects on colonic health influence the efficacy of Bin1 mAb immunotherapy for ulcerative colitis. Sci Rep 2023; 13:11802. [PMID: 37479833 PMCID: PMC10361997 DOI: 10.1038/s41598-023-38830-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/16/2023] [Indexed: 07/23/2023] Open
Abstract
Ulcerative colitis (UC) is an idiopathic disease of the large intestine linked to high fat-high protein diets, a dysbiotic microbiome, and a metabolome linked to diet and/or aberrant circadian rhythms associated with poor sleeping patterns. Understanding diet-affected factors that negatively influence colonic health may offer new insights into how to prevent UC and enhance the efficacy of UC immunotherapy. In this preclinical study, we found that standard or high fiber diets in mice positively influenced their colonic health, whereas a high fat-high protein diet negatively influenced colonic health, consistent with clinical findings. Animals fed a high fat/high protein diet experienced obesity and a reduced colon length, illustrating a phenotype we suggest calling peinosis [hunger-like-condition; Greek, peina: hunger; osis: condition], as marked by a lack of nutrient energy remaining in fecal pellets. Notably, a high fat/high protein diet also led to signs of muscle weakness that could not be explained fully by weight gain. In contrast, mice on a high fiber diet ranked highest compared to other diets in terms of colon length and lack of muscle weakness. That said, mice on a high fiber diet were more prone to UC and toxic responses to immunotherapy, consistent with clinical observations. Recent studies have suggested that a standard diet may be needed to support the efficacy of immunotherapeutic drugs used to prevent and treat UC. Here we observed that protection against UC by Bin1 mAb, a passive UC immunotherapy that acts by coordinately enforcing intestinal barrier function, protecting enteric neurons, and normalizing the microbiome, was associated with increased colonic levels of healthful short-chain fatty acids (SCFA), particularly butyric acid and propionic acid, which help enforce intestinal barrier function. This work offers a preclinical platform to investigate how diet affects UC immunotherapy and the potential of dietary SCFA supplements to enhance it. Further, it suggests that the beneficial effects of passive immunotherapy by Bin1 mAb in UC treatment may be mediated to some extent by promoting increased levels of healthful SCFA.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA.
| | - Nickey Dilbarov
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Joseph Kelly
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA
| | | | - George C Prendergast
- Lankenau Institute for Medical Research, 100 E. Lancaster Avenue, Wynnewood, PA, 19096, USA
| |
Collapse
|
4
|
Thomas S, Prendergast GC. Gut-brain connections in neurodegenerative disease: immunotherapeutic targeting of Bin1 in inflammatory bowel disease and Alzheimer's disease. Front Pharmacol 2023; 14:1183932. [PMID: 37521457 PMCID: PMC10372349 DOI: 10.3389/fphar.2023.1183932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023] Open
Abstract
Longer lifespan produces risks of age-associated neurodegenerative disorders such as Alzheimer's disease (AD), which is characterized by declines in memory and cognitive function. The pathogenic causes of AD are thought to reflect a progressive aggregation in the brain of amyloid plaques composed of beta-amyloid (Aß) peptides and neurofibrillary tangles composed of phosphorylated tau protein. Recently, long-standing investigations of the Aß disease hypothesis gained support via a passive immunotherapy targeting soluble Aß protein. Tau-targeting approaches using antibodies are also being pursued as a therapeutic approach to AD. In genome-wide association studies, the disease modifier gene Bin1 has been identified as a top risk factor for late-onset AD in human populations, with recent studies suggesting that Bin1 binds tau and influences its extracellular deposition. Interestingly, before AD emerges in the brain, tau levels rise in the colon, where Bin1-a modifier of tissue barrier function and inflammation-acts to promote inflammatory bowel disease (IBD). This connection is provocative given clinical evidence of gut-brain communication in age-associated neurodegenerative disorders, including AD. In this review, we discuss a Bin1-targeting passive immunotherapy developed in our laboratory to treat IBD that may offer a strategy to indirectly reduce tau deposition and limit AD onset or progression.
Collapse
|
5
|
Ancuceanu R, Hovaneț MV, Cojocaru-Toma M, Anghel AI, Dinu M. Potential Antifungal Targets for Aspergillus sp. from the Calcineurin and Heat Shock Protein Pathways. Int J Mol Sci 2022; 23:ijms232012543. [PMID: 36293395 PMCID: PMC9603945 DOI: 10.3390/ijms232012543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Aspergillus species, especially A. fumigatus, and to a lesser extent others (A. flavus, A. niger, A. terreus), although rarely pathogenic to healthy humans, can be very aggressive to immunocompromised patients (they are opportunistic pathogens). Although survival rates for such infections have improved in recent decades following the introduction of azole derivatives, they remain a clinical challenge. The fact that current antifungals act as fungistatic rather than fungicide, that they have limited safety, and that resistance is becoming increasingly common make the need for new, more effective, and safer therapies to become more acute. Over the last decades, knowledge about the molecular biology of A. fumigatus and other Aspergillus species, and particularly of calcineurin, Hsp90, and their signaling pathway proteins, has progressed remarkably. Although calcineurin has attracted much interest, its adverse effects, particularly its immunosuppressive effects, make it less attractive than it might at first appear. The situation is not very different for Hsp90. Other proteins from their signaling pathways, such as protein kinases phosphorylating the four SPRR serine residues, CrzA, rcnA, pmcA-pmcC (particularly pmcC), rfeF, BAR adapter protein(s), the phkB histidine kinase, sskB MAP kinase kinase, zfpA, htfA, ctfA, SwoH (nucleoside diphosphate kinase), CchA, MidA, FKBP12, the K27 lysine position from Hsp90, PkcA, MpkA, RlmA, brlA, abaA, wetA, other heat shock proteins (Hsp70, Hsp40, Hsp12) currently appear promising and deserve further investigation as potential targets for antifungal drug development.
Collapse
Affiliation(s)
- Robert Ancuceanu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
- Correspondence: (R.A.); (M.V.H.)
| | - Marilena Viorica Hovaneț
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
- Correspondence: (R.A.); (M.V.H.)
| | - Maria Cojocaru-Toma
- Faculty of Pharmacy, Nicolae Testemițanu State University of Medicine and Pharmacy, 2025 Chisinau, Moldova
| | - Adriana-Iuliana Anghel
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Mihaela Dinu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
6
|
Guo G, Gong K, Beckley N, Zhang Y, Yang X, Chkheidze R, Hatanpaa KJ, Garzon-Muvdi T, Koduru P, Nayab A, Jenks J, Sathe AA, Liu Y, Xing C, Wu SY, Chiang CM, Mukherjee B, Burma S, Wohlfeld B, Patel T, Mickey B, Abdullah K, Youssef M, Pan E, Gerber DE, Tian S, Sarkaria JN, McBrayer SK, Zhao D, Habib AA. EGFR ligand shifts the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastoma by suppressing invasion through BIN3 upregulation. Nat Cell Biol 2022; 24:1291-1305. [PMID: 35915159 PMCID: PMC9389625 DOI: 10.1038/s41556-022-00962-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 06/14/2022] [Indexed: 02/03/2023]
Abstract
The epidermal growth factor receptor (EGFR) is a prime oncogene that is frequently amplified in glioblastomas. Here we demonstrate a new tumour-suppressive function of EGFR in EGFR-amplified glioblastomas regulated by EGFR ligands. Constitutive EGFR signalling promotes invasion via activation of a TAB1-TAK1-NF-κB-EMP1 pathway, resulting in large tumours and decreased survival in orthotopic models. Ligand-activated EGFR promotes proliferation and surprisingly suppresses invasion by upregulating BIN3, which inhibits a DOCK7-regulated Rho GTPase pathway, resulting in small hyperproliferating non-invasive tumours and improved survival. Data from The Cancer Genome Atlas reveal that in EGFR-amplified glioblastomas, a low level of EGFR ligands confers a worse prognosis, whereas a high level of EGFR ligands confers an improved prognosis. Thus, increased EGFR ligand levels shift the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastomas by suppressing invasion. The tumour-suppressive function of EGFR can be activated therapeutically using tofacitinib, which suppresses invasion by increasing EGFR ligand levels and upregulating BIN3.
Collapse
Affiliation(s)
- Gao Guo
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ke Gong
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, School of Basic Medical Sciences, Taikang Medical School, Wuhan University, Wuhan, China
| | - Nicole Beckley
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yue Zhang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoyao Yang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rati Chkheidze
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kimmo J Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prasad Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arifa Nayab
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jennifer Jenks
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adwait Amod Sathe
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Liu
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shwu-Yuan Wu
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharamacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheng-Ming Chiang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharamacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Bryan Wohlfeld
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Toral Patel
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce Mickey
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kalil Abdullah
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Youssef
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward Pan
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David E Gerber
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Hematology-Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shulan Tian
- Department of Quantitative Heath Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Samuel K McBrayer
- Department of Pediatrics and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dawen Zhao
- Departments of Biomedical Engineering and Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Amyn A Habib
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- VA North Texas Health Care System, Dallas, TX, USA.
| |
Collapse
|
7
|
Taga M, Petyuk VA, White C, Marsh G, Ma Y, Klein HU, Connor SM, Kroshilina A, Yung CJ, Khairallah A, Olah M, Schneider J, Karhohs K, Carpenter AE, Ransohoff R, Bennett DA, Crotti A, Bradshaw EM, De Jager PL. BIN1 protein isoforms are differentially expressed in astrocytes, neurons, and microglia: neuronal and astrocyte BIN1 are implicated in tau pathology. Mol Neurodegener 2020; 15:44. [PMID: 32727516 PMCID: PMC7389646 DOI: 10.1186/s13024-020-00387-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 06/08/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Identified as an Alzheimer's disease (AD) susceptibility gene by genome wide-association studies, BIN1 has 10 isoforms that are expressed in the Central Nervous System (CNS). The distribution of these isoforms in different cell types, as well as their role in AD pathology still remains unclear. METHODS Utilizing antibodies targeting specific BIN1 epitopes in human post-mortem tissue and analyzing mRNA expression data from purified microglia, we identified three isoforms expressed in neurons and astrocytes (isoforms 1, 2 and 3) and four isoforms expressed in microglia (isoforms 6, 9, 10 and 12). The abundance of selected peptides, which correspond to groups of BIN1 protein isoforms, was measured in dorsolateral prefrontal cortex, and their relation to neuropathological features of AD was assessed. RESULTS Peptides contained in exon 7 of BIN1's N-BAR domain were found to be significantly associated with AD-related traits and, particularly, tau tangles. Decreased expression of BIN1 isoforms containing exon 7 is associated with greater accumulation of tangles and subsequent cognitive decline, with astrocytic rather than neuronal BIN1 being the more likely culprit. These effects are independent of the BIN1 AD risk variant. CONCLUSIONS Exploring the molecular mechanisms of specific BIN1 isoforms expressed by astrocytes may open new avenues for modulating the accumulation of Tau pathology in AD.
Collapse
Affiliation(s)
- Mariko Taga
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | | | - Charles White
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | | | - Yiyi Ma
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | - Sarah M. Connor
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | - Alexandra Kroshilina
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
| | - Christina J. Yung
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
| | - Anthony Khairallah
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
| | - Marta Olah
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | - Julie Schneider
- Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Kyle Karhohs
- Imaging Platform, Broad Institute, Cambridge, MA USA
| | | | - Richard Ransohoff
- Third Rock Ventures, 29 Newbury Street, Suite 301, Boston, MA 02116 USA
- Department of Cell Biology, Harvard Medical School, Boston, MA USA
| | - David A. Bennett
- Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | | | - Elizabeth M. Bradshaw
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th st, PH19-311, New York, NY 10032 USA
- Cell Circuits Program, Broad Institute, Cambridge, MA USA
| |
Collapse
|
8
|
Le Naour J, Galluzzi L, Zitvogel L, Kroemer G, Vacchelli E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2020; 9:1777625. [PMID: 32934882 PMCID: PMC7466863 DOI: 10.1080/2162402x.2020.1777625] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first, rate-limiting step of the so-called “kynurenine pathway”, which converts the essential amino acid L-tryptophan (Trp) into the immunosuppressive metabolite L-kynurenine (Kyn). While expressed constitutively by some tissues, IDO1 can also be induced in specific subsets of antigen-presenting cells that ultimately favor the establishment of immune tolerance to tumor antigens. At least in part, the immunomodulatory functions of IDO1 can be explained by depletion of Trp and accumulation of Kyn and its derivatives. In animal tumor models, genetic or pharmacological IDO1 inhibition can cause the (re)activation of anticancer immune responses. Similarly, neoplasms expressing high levels of IDO1 may elude anticancer immunosurveillance. Therefore, IDO1 inhibitors represent promising therapeutic candidates for cancer therapy, and some of them have already entered clinical evaluation. Here, we summarize preclinical and clinical studies testing IDO1-targeting interventions for oncologic indications.
Collapse
Affiliation(s)
- Julie Le Naour
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université De Paris, Sorbonne Université, INSERM U1138, Centre De Recherche Des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine Kremlin Bicêtre, Université Paris Sud, Paris Saclay, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université De Paris, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France.,Equipe Labellisée Ligue Contre Le Cancer, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université De Paris, Sorbonne Université, INSERM U1138, Centre De Recherche Des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Erika Vacchelli
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université De Paris, Sorbonne Université, INSERM U1138, Centre De Recherche Des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
9
|
McAvoy KM, Rajamohamed Sait H, Marsh G, Peterson M, Reynolds TL, Gagnon J, Geisler S, Leach P, Roberts C, Cahir-McFarland E, Ransohoff RM, Crotti A. Cell-autonomous and non-cell autonomous effects of neuronal BIN1 loss in vivo. PLoS One 2019; 14:e0220125. [PMID: 31408457 PMCID: PMC6692034 DOI: 10.1371/journal.pone.0220125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/09/2019] [Indexed: 01/25/2023] Open
Abstract
BIN1 is the most important risk locus for Late Onset Alzheimer’s Disease (LOAD), after ApoE. BIN1 AD-associated SNPs correlate with Tau deposition as well as with brain atrophy. Furthermore, the level of neuronal-specific BIN1 isoform 1 protein is decreased in sporadic AD cases in parallel with neuronal loss, despite an overall increase in BIN1 total mRNA. To address the relationship between reduction of BIN1 and neuronal cell loss in the context of Tau pathology, we knocked-down endogenous murine Bin1 via stereotaxic injection of AAV-Bin1 shRNA in the hippocampus of mice expressing Tau P301S (PS19). We observed a statistically significant reduction in the number of neurons in the hippocampus of mice injected with AAV-Bin1 shRNA in comparison with mice injected with AAV control. To investigate whether neuronal loss is due to deletion of Bin1 selectively in neurons in presence Tau P301S, we bred Bin1flox/flox with Thy1-Cre and subsequently with PS19 mice. Mice lacking neuronal Bin1 and expressing Tau P301S showed increased mortality, without increased neuropathology, when compared to neuronal Bin1 and Tau P301S-expressing mice. The loss of Bin1 isoform 1 resulted in reduced excitability in primary neurons in vitro, reduced neuronal c-fos expression as well as in altered microglia transcriptome in vivo. Taken together, our data suggest that the contribution of genetic variation in BIN1 locus to AD risk could result from a cell-autonomous reduction of neuronal excitability due to Bin1 decrease, exacerbated by the presence of aggregated Tau, coupled with a non-cell autonomous microglia activation.
Collapse
Affiliation(s)
| | | | | | | | | | - Jake Gagnon
- Biogen, Cambridge, MA, United States of America
| | | | | | | | | | - Richard M Ransohoff
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States of America
| | | |
Collapse
|
10
|
Crotti A, Sait HR, McAvoy KM, Estrada K, Ergun A, Szak S, Marsh G, Jandreski L, Peterson M, Reynolds TL, Dalkilic-Liddle I, Cameron A, Cahir-McFarland E, Ransohoff RM. BIN1 favors the spreading of Tau via extracellular vesicles. Sci Rep 2019; 9:9477. [PMID: 31263146 PMCID: PMC6603165 DOI: 10.1038/s41598-019-45676-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/06/2019] [Indexed: 01/09/2023] Open
Abstract
Despite Bridging INtegrator 1 (BIN1) being the second most statistically-significant locus associated to Late Onset Alzheimer’s Disease, its role in disease pathogenesis remains to be clarified. As reports suggest a link between BIN1, Tau and extracellular vesicles, we investigated whether BIN1 could affect Tau spreading via exosomes secretion. We observed that BIN1-associated Tau-containing extracellular vesicles purified from cerebrospinal fluid of AD-affected individuals are seeding-competent. We showed that BIN1 over-expression promotes the release of Tau via extracellular vesicles in vitro as well as exacerbation of Tau pathology in vivo in PS19 mice. Genetic deletion of Bin1 from microglia resulted in reduction of Tau secretion via extracellular vesicles in vitro, and in decrease of Tau spreading in vivo in male, but not female, mice, in the context of PS19 background. Interestingly, ablation of Bin1 in microglia of male mice resulted in significant reduction in the expression of heat-shock proteins, previously implicated in Tau proteostasis. These observations suggest that BIN1 could contribute to the progression of AD-related Tau pathology by altering Tau clearance and promoting release of Tau-enriched extracellular vesicles by microglia.
Collapse
Affiliation(s)
- Andrea Crotti
- Biogen, 225 Binney St., Cambridge, MA, 02142, USA. .,Astellas, 1030 Massachusetts Avenue, Cambridge, MA, 02138, USA.
| | | | | | | | - Ayla Ergun
- Fulcrum Therapeutics, 26 Landsdowne St, Cambridge, MA, 02139, USA
| | - Suzanne Szak
- Biogen, 225 Binney St., Cambridge, MA, 02142, USA
| | - Galina Marsh
- Biogen, 225 Binney St., Cambridge, MA, 02142, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Fox E, Oliver T, Rowe M, Thomas S, Zakharia Y, Gilman PB, Muller AJ, Prendergast GC. Indoximod: An Immunometabolic Adjuvant That Empowers T Cell Activity in Cancer. Front Oncol 2018; 8:370. [PMID: 30254983 PMCID: PMC6141803 DOI: 10.3389/fonc.2018.00370] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/21/2018] [Indexed: 11/20/2022] Open
Abstract
Exploding interest in immunometabolism as a source of new cancer therapeutics has been driven in large part by studies of tryptophan catabolism mediated by IDO/TDO enzymes. A chief focus in the field is IDO1, a pro-inflammatory modifier that is widely overexpressed in cancers where it blunts immunosurveillance and enables neovascularization and metastasis. The simple racemic compound 1-methyl-D,L-tryptophan (1MT) is an extensively used probe of IDO/TDO pathways that exerts a variety of complex inhibitory effects. The L isomer of 1MT is a weak substrate for IDO1 and is ascribed the weak inhibitory activity of the racemate on the enzyme. In contrast, the D isomer neither binds nor inhibits the purified IDO1 enzyme. However, clinical development focused on D-1MT (now termed indoximod) due to preclinical cues of its greater anticancer activity and its distinct mechanisms of action. In contrast to direct enzymatic inhibitors of IDO1, indoximod acts downstream of IDO1 to stimulate mTORC1, a convergent effector signaling molecule for all IDO/TDO enzymes, thus possibly lowering risks of drug resistance by IDO1 bypass. In this review, we survey the unique biological and mechanistic features of indoximod as an IDO/TDO pathway inhibitor, including recent clinical findings of its ability to safely enhance various types of cancer therapy, including chemotherapy, chemo-radiotherapy, vaccines, and immune checkpoint therapy. We also review the potential advantages indoximod offers compared to selective IDO1-specific blockade, which preclinical studies and the clinical study ECHO-301 suggest may be bypassed readily by tumors. Indoximod lies at a leading edge of broad-spectrum immunometabolic agents that may act to improve responses to many anticancer modalities, in a manner analogous to vaccine adjuvants that act to boost immunity in settings of infectious disease.
Collapse
Affiliation(s)
- Eric Fox
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Thomas Oliver
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Melissa Rowe
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
| | - Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Yousef Zakharia
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Paul B. Gilman
- Department of Hematology-Oncology, Lankenau Medical Center, Wynnewood, PA, United States
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Alexander J. Muller
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
12
|
Alternative Splicing as a Target for Cancer Treatment. Int J Mol Sci 2018; 19:ijms19020545. [PMID: 29439487 PMCID: PMC5855767 DOI: 10.3390/ijms19020545] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 02/06/2023] Open
Abstract
Alternative splicing is a key mechanism determinant for gene expression in metazoan. During alternative splicing, non-coding sequences are removed to generate different mature messenger RNAs due to a combination of sequence elements and cellular factors that contribute to splicing regulation. A different combination of splicing sites, exonic or intronic sequences, mutually exclusive exons or retained introns could be selected during alternative splicing to generate different mature mRNAs that could in turn produce distinct protein products. Alternative splicing is the main source of protein diversity responsible for 90% of human gene expression, and it has recently become a hallmark for cancer with a full potential as a prognostic and therapeutic tool. Currently, more than 15,000 alternative splicing events have been associated to different aspects of cancer biology, including cell proliferation and invasion, apoptosis resistance and susceptibility to different chemotherapeutic drugs. Here, we present well established and newly discovered splicing events that occur in different cancer-related genes, their modification by several approaches and the current status of key tools developed to target alternative splicing with diagnostic and therapeutic purposes.
Collapse
|
13
|
Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ. Discovery of IDO1 Inhibitors: From Bench to Bedside. Cancer Res 2018; 77:6795-6811. [PMID: 29247038 DOI: 10.1158/0008-5472.can-17-2285] [Citation(s) in RCA: 412] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/23/2017] [Accepted: 11/01/2017] [Indexed: 01/11/2023]
Abstract
Small-molecule inhibitors of indoleamine 2,3-dioxygenase-1 (IDO1) are emerging at the vanguard of experimental agents in oncology. Here, pioneers of this new drug class provide a bench-to-bedside review on preclinical validation of IDO1 as a cancer therapeutic target and on the discovery and development of a set of mechanistically distinct compounds, indoximod, epacadostat, and navoximod, that were first to be evaluated as IDO inhibitors in clinical trials. As immunometabolic adjuvants to widen therapeutic windows, IDO inhibitors may leverage not only immuno-oncology modalities but also chemotherapy and radiotherapy as standards of care in the oncology clinic. Cancer Res; 77(24); 6795-811. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - James B DuHadaway
- Lankenau Institute for Medical Research (LIMR), Wynnewood, Pennsylvania
| | | |
Collapse
|
14
|
Inflammatory Reprogramming with IDO1 Inhibitors: Turning Immunologically Unresponsive 'Cold' Tumors 'Hot'. Trends Cancer 2017; 4:38-58. [PMID: 29413421 DOI: 10.1016/j.trecan.2017.11.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 01/24/2023]
Abstract
We discuss how small-molecule inhibitors of the tryptophan (Trp) catabolic enzyme indoleamine 2,3-dioxygenase (IDO) represent a vanguard of new immunometabolic adjuvants to safely enhance the efficacy of cancer immunotherapy, radiotherapy, or 'immunogenic' chemotherapy by leveraging responses to tumor neoantigens. IDO inhibitors re-program inflammatory processes to help clear tumors by blunting tumor neovascularization and restoring immunosurveillance. Studies of regulatory and effector pathways illuminate IDO as an inflammatory modifier. Recent work suggests that coordinate targeting of the Trp catabolic enzymes tryptophan 2,3-dioxygenase (TDO) and IDO2 may also safely broaden efficacy. Understanding IDO inhibitors as adjuvants to turn immunologically 'cold' tumors 'hot' can seed new concepts in how to improve the efficacy of cancer therapy while limiting collateral damage.
Collapse
|
15
|
Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 336:175-203. [PMID: 29413890 PMCID: PMC6054468 DOI: 10.1016/bs.ircmb.2017.07.004] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The tryptophan catabolic enzyme indoleamine 2,3-dioxygenase-1 (IDO1) has attracted enormous attention in driving cancer immunosuppression, neovascularization, and metastasis. IDO1 suppresses local CD8+ T effector cells and natural killer cells and induces CD4+ T regulatory cells (iTreg) and myeloid-derived suppressor cells (MDSC). The structurally distinct enzyme tryptophan dioxygenase (TDO) also has been implicated recently in immune escape and metastatic progression. Lastly, emerging evidence suggests that the IDO1-related enzyme IDO2 may support IDO1-mediated iTreg and contribute to B-cell inflammed states in certain cancers. IDO1 and TDO are upregulated widely in neoplastic cells but also variably in stromal, endothelial, and innate immune cells of the tumor microenviroment and in tumor-draining lymph nodes. Pharmacological and genetic proofs in preclinical models of cancer have validated IDO1 as a cancer therapeutic target. IDO1 inhibitors have limited activity on their own but greatly enhance "immunogenic" chemotherapy or immune checkpoint drugs. IDO/TDO function is rooted in inflammatory programming, thereby influencing tumor neovascularization, MDSC generation, and metastasis beyond effects on adaptive immune tolerance. Discovery and development of two small molecule enzyme inhibitors of IDO1 have advanced furthest to date in Phase II/III human trials (epacadostat and navoximod, respectively). Indoximod, a tryptophan mimetic compound with a different mechanism of action in the IDO pathway has also advanced in multiple Phase II trials. Second generation combined IDO/TDO inhibitors may broaden impact in cancer treatment, for example, in addressing IDO1 bypass (inherent resistance) or acquired resistance to IDO1 inhibitors. This review surveys knowledge about IDO1 function and how IDO1 inhibitors reprogram inflammation to heighten therapeutic responses in cancer.
Collapse
Affiliation(s)
- George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| | | | - Arpita Mondal
- Lankenau Institute for Medical Research, Wynnewood, PA, United States; Drexel University College of Medicine, Philadelphia, PA, United States
| | - Peggy Scherle
- Incyte Corporation Inc., Wilmington, DE, United States
| | - Alexander J Muller
- Lankenau Institute for Medical Research, Wynnewood, PA, United States; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
16
|
De Rossi P, Buggia-Prévot V, Clayton BLL, Vasquez JB, van Sanford C, Andrew RJ, Lesnick R, Botté A, Deyts C, Salem S, Rao E, Rice RC, Parent A, Kar S, Popko B, Pytel P, Estus S, Thinakaran G. Predominant expression of Alzheimer's disease-associated BIN1 in mature oligodendrocytes and localization to white matter tracts. Mol Neurodegener 2016; 11:59. [PMID: 27488240 PMCID: PMC4973113 DOI: 10.1186/s13024-016-0124-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/27/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Genome-wide association studies have identified BIN1 within the second most significant susceptibility locus in late-onset Alzheimer's disease (AD). BIN1 undergoes complex alternative splicing to generate multiple isoforms with diverse functions in multiple cellular processes including endocytosis and membrane remodeling. An increase in BIN1 expression in AD and an interaction between BIN1 and Tau have been reported. However, disparate descriptions of BIN1 expression and localization in the brain previously reported in the literature and the lack of clarity on brain BIN1 isoforms present formidable challenges to our understanding of how genetic variants in BIN1 increase the risk for AD. METHODS In this study, we analyzed BIN1 mRNA and protein levels in human brain samples from individuals with or without AD. In addition, we characterized the BIN1 expression and isoform diversity in human and rodent tissue by immunohistochemistry and immunoblotting using a panel of BIN1 antibodies. RESULTS Here, we report on BIN1 isoform diversity in the human brain and document alterations in the levels of select BIN1 isoforms in individuals with AD. In addition, we report striking BIN1 localization to white matter tracts in rodent and the human brain, and document that the large majority of BIN1 is expressed in mature oligodendrocytes whereas neuronal BIN1 represents a minor fraction. This predominant non-neuronal BIN1 localization contrasts with the strict neuronal expression and presynaptic localization of the BIN1 paralog, Amphiphysin 1. We also observe upregulation of BIN1 at the onset of postnatal myelination in the brain and during differentiation of cultured oligodendrocytes. Finally, we document that the loss of BIN1 significantly correlates with the extent of demyelination in multiple sclerosis lesions. CONCLUSION Our study provides new insights into the brain distribution and cellular expression of an important risk factor associated with late-onset AD. We propose that efforts to define how genetic variants in BIN1 elevate the risk for AD would behoove to consider BIN1 function in the context of its main expression in mature oligodendrocytes and the potential for a role of BIN1 in the membrane remodeling that accompanies the process of myelination.
Collapse
Affiliation(s)
- Pierre De Rossi
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Virginie Buggia-Prévot
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | | | - Jared B. Vasquez
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Carson van Sanford
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Robert J. Andrew
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Ruben Lesnick
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Alexandra Botté
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Carole Deyts
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Someya Salem
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Eshaan Rao
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Richard C. Rice
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Angèle Parent
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Satyabrata Kar
- Centre for prions and protein folding diseases, University of Alberta, Edmonton, AB T6G 2B7 Canada
| | - Brian Popko
- Department of Neurology, The University of Chicago, Chicago, IL 60637 USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL 60637 USA
| | - Steven Estus
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
- Department of Neurology, The University of Chicago, Chicago, IL 60637 USA
- Department of Pathology, The University of Chicago, Chicago, IL 60637 USA
| |
Collapse
|
17
|
Pyndiah S, Sakamuro D. Restoration of tumor suppressor functions by small-molecule inhibitors. Mol Cell Oncol 2016; 2:e991225. [PMID: 27308472 PMCID: PMC4905308 DOI: 10.4161/23723556.2014.991225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 11/20/2014] [Accepted: 11/20/2014] [Indexed: 11/24/2022]
Abstract
Over the last decades, accumulating data have advanced our understanding of the mechanism of action of tumor suppressor proteins and therapeutic strategies to restore tumor suppressor pathways have emerged as a promising approach for cancer therapy. Based on our recent findings on bridging integrator-1 (BIN1), we outline potential advantages and disadvantages of chemical activation of tumor suppressors.
Collapse
Affiliation(s)
| | - Daitoku Sakamuro
- Department of Biochemistry and Molecular Biology; Medical College of Georgia; Georgia Regents University Cancer Center ; Augusta, GA, USA
| |
Collapse
|
18
|
Laury-Kleintop LD, Mulgrew JR, Heletz I, Nedelcoviciu RA, Chang MY, Harris DM, Koch WJ, Schneider MD, Muller AJ, Prendergast GC. Cardiac-specific disruption of Bin1 in mice enables a model of stress- and age-associated dilated cardiomyopathy. J Cell Biochem 2016; 116:2541-51. [PMID: 25939245 DOI: 10.1002/jcb.25198] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/14/2015] [Indexed: 12/21/2022]
Abstract
Non-compensated dilated cardiomyopathy (DCM) leading to death from heart failure is rising rapidly in developed countries due to aging demographics, and there is a need for informative preclinical models to guide the development of effective therapeutic strategies to prevent or delay disease onset. In this study, we describe a novel model of heart failure based on cardiac-specific deletion of the prototypical mammalian BAR adapter-encoding gene Bin1, a modifier of age-associated disease. Bin1 deletion during embryonic development causes hypertrophic cardiomyopathy and neonatal lethality, but there is little information on how Bin1 affects cardiac function in adult animals. Here we report that cardiomyocyte-specific loss of Bin1 causes age-associated dilated cardiomyopathy (DCM) beginning by 8-10 months of age. Echocardiographic analysis showed that Bin1 loss caused a 45% reduction in ejection fraction during aging. Younger animals rapidly developed DCM if cardiac pressure overload was created by transverse aortic constriction. Heterozygotes exhibited an intermediate phenotype indicating Bin1 is haplo-insufficient to sustain normal heart function. Bin1 loss increased left ventricle (LV) volume and diameter during aging, but it did not alter LV volume or diameter in hearts from heterozygous mice nor did it affect LV mass. Bin1 loss increased interstitial fibrosis and mislocalization of the voltage-dependent calcium channel Cav 1.2, and the lipid raft scaffold protein caveolin-3, which normally complexes with Bin1 and Cav 1.2 in cardiomyocyte membranes. Our findings show how cardiac deficiency in Bin1 function causes age- and stress-associated heart failure, and they establish a new preclinical model of this terminal cardiac disease.
Collapse
Affiliation(s)
| | | | - Ido Heletz
- Lankenau Medical Center, Wynnewood, Pennsylvania
| | | | - Mee Young Chang
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - David M Harris
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine, Temple University Medical School, Philadelphia, Pennsylvania
| | - Michael D Schneider
- National Heart and Lung Institute, British Heart Foundation Centre of Research Excellence, Faculty of Medicine, Imperial College London, London, UK
| | | | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.,Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical School and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Thomas S, Mercado JM, DuHadaway J, DiGuilio K, Mullin JM, Prendergast GC. Novel Colitis Immunotherapy Targets Bin1 and Improves Colon Cell Barrier Function. Dig Dis Sci 2016. [PMID: 26195312 DOI: 10.1007/s10620-015-3804-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Ulcerative colitis (UC) is associated with defects in colonic epithelial barriers as well as inflammation of the colon mucosa resulting from the recruitment of lymphocytes and neutrophils in the lamina propria. Patients afflicted with UC are at increased risk of colorectal cancer. Currently, UC management employs general anti-inflammatory strategies associated with a variety of side effects, including heightened risks of infection, in patients where the therapy is variably effective. Thus, second generation drugs that can more effectively and selectively limit UC are desired. AIM Building on genetic evidence that attenuation of the Bin1 (Bridging integrator 1) gene can limit UC pathogenicity in the mouse, we pursued Bin1 targeting as a therapeutic option. METHODS Mice were injected with a single dose of Bin1 mAb followed by oral administration of 3 % DSS in water for 7 days. RESULTS In this study, we offer preclinical proof of concept for a monoclonal antibody (mAb) targeting the Bin1 protein that blunts UC pathogenicity in a mouse model of experimental colitis. Administration of Bin1 mAb reduced colitis morbidity in mice; whereas unprotected mice is characterized by severe lesions throughout the mucosa, rupture of the lymphoid follicle, high-level neutrophil and lymphocyte infiltration into the mucosal and submucosal areas, and loss of surface crypts. In vitro studies in human Caco-2 cells showed that Bin1 antibody altered the expression of tight junction proteins and improved barrier function. CONCLUSIONS Our results suggest that a therapy based on Bin1 monoclonal antibody supporting mucosal barrier function and protecting integrity of the lymphoid follicle could offer a novel strategy to treat UC and possibly limit risks of colorectal cancer.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.
| | - Joanna M Mercado
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.,Division of Gastroenterology, Lankenau Medical Center, Wynnewood, PA, 19096, USA
| | - James DuHadaway
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Kate DiGuilio
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.,Division of Gastroenterology, Lankenau Medical Center, Wynnewood, PA, 19096, USA
| | - James M Mullin
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.,Division of Gastroenterology, Lankenau Medical Center, Wynnewood, PA, 19096, USA
| | - George C Prendergast
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.,Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical School, Philadelphia, PA, 19107, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| |
Collapse
|
20
|
Splicing Regulators and Their Roles in Cancer Biology and Therapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:150514. [PMID: 26273588 PMCID: PMC4529883 DOI: 10.1155/2015/150514] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/10/2015] [Accepted: 04/01/2015] [Indexed: 12/17/2022]
Abstract
Alternative splicing allows cells to expand the encoding potential of their genomes. In this elegant mechanism, a single gene can yield protein isoforms with even antagonistic functions depending on the cellular physiological context. Alterations in splicing regulatory factors activity in cancer cells, however, can generate an abnormal protein expression pattern that promotes growth, survival, and other processes, which are relevant to tumor biology. In this review, we discuss dysregulated alternative splicing events and regulatory factors that impact pathways related to cancer. The SR proteins and their regulatory kinases SRPKs and CLKs have been frequently found altered in tumors and are examined in more detail. Finally, perspectives that support splicing machinery as target for the development of novel anticancer therapies are discussed.
Collapse
|
21
|
Abstract
BAR proteins comprise a heterogeneous group of multi-domain proteins with diverse biological functions. The common denominator is the Bin-Amphiphysin-Rvs (BAR) domain that not only confers targeting to lipid bilayers, but also provides scaffolding to mold lipid membranes into concave or convex surfaces. This function of BAR proteins is an important determinant in the dynamic reconstruction of membrane vesicles, as well as of the plasma membrane. Several BAR proteins function as linkers between cytoskeletal regulation and membrane dynamics. These links are provided by direct interactions between BAR proteins and actin-nucleation-promoting factors of the Wiskott-Aldrich syndrome protein family and the Diaphanous-related formins. The Rho GTPases are key factors for orchestration of this intricate interplay. This review describes how BAR proteins regulate the activity of Rho GTPases, as well as how Rho GTPases regulate the function of BAR proteins. This mutual collaboration is a central factor in the regulation of vital cellular processes, such as cell migration, cytokinesis, intracellular transport, endocytosis, and exocytosis.
Collapse
Affiliation(s)
- Pontus Aspenström
- a Department of Microbiology and Tumor and Cell Biology; Karolinska Institutet ; Stockholm , Sweden
| |
Collapse
|
22
|
Kumari A, Iwasaki T, Pyndiah S, Cassimere EK, Palani CD, Sakamuro D. Regulation of E2F1-induced apoptosis by poly(ADP-ribosyl)ation. Cell Death Differ 2014; 22:311-22. [PMID: 25257171 DOI: 10.1038/cdd.2014.146] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/16/2014] [Accepted: 08/18/2014] [Indexed: 11/09/2022] Open
Abstract
The transcription factor adenovirus E2 promoter-binding factor (E2F)-1 normally enhances cell-cycle progression, but it also induces apoptosis under certain conditions, including DNA damage and serum deprivation. Although DNA damage facilitates the phosphorylation and stabilization of E2F1 to trigger apoptosis, how serum starvation renders cells vulnerable to E2F1-induced apoptosis remains unclear. Because poly(ADP-ribose) polymerase 1 (PARP1), a nuclear enzyme essential for genomic stability and chromatin remodeling, interacts directly with E2F1, we investigated the effects of PARP1 on E2F1-mediated functions in the presence and absence of serum. PARP1 attenuation, which increased E2F1 transactivation, induced G2/M cell-cycle arrest under normal growth conditions, but enhanced E2F1-induced apoptosis in serum-starved cells. Interestingly, basal PARP1 activity was sufficient to modify E2F1 by poly(ADP-ribosyl)ation, which stabilized the interaction between E2F1 and the BIN1 tumor suppressor in the nucleus. Accordingly, BIN1 acted as an RB1-independent E2F1 corepressor. Because E2F1 directly activates the BIN1 gene promoter, BIN1 curbed E2F1 activity through a negative-feedback mechanism. Conversely, when the BIN1-E2F1 interaction was abolished by PARP1 suppression, E2F1 continuously increased BIN1 levels. This is functionally germane, as PARP1-depletion-associated G2/M arrest was reversed by the transfection of BIN1 siRNA. Moreover, PARP-inhibitor-associated anti-transformation activity was compromised by the coexpression of dominant-negative BIN1. Because serum starvation massively reduced the E2F1 poly(ADP-ribosyl)ation, we conclude that the release of BIN1 from hypo-poly(ADP-ribosyl)ated E2F1 is a mechanism by which serum starvation promotes E2F1-induced apoptosis.
Collapse
Affiliation(s)
- A Kumari
- 1] Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University Cancer Center, Augusta, GA 30912, USA [2] Molecular Signaling Program, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - T Iwasaki
- 1] Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University Cancer Center, Augusta, GA 30912, USA [2] Laboratory of Molecular Biology, Research Center for Environmental Genomics, Kobe University, Kobe 657, Japan
| | - S Pyndiah
- Molecular Signaling Program, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - E K Cassimere
- Molecular Signaling Program, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - C D Palani
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| | - D Sakamuro
- 1] Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University Cancer Center, Augusta, GA 30912, USA [2] Molecular Signaling Program, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
23
|
Wu T, Shi Z, Baumgart T. Mutations in BIN1 associated with centronuclear myopathy disrupt membrane remodeling by affecting protein density and oligomerization. PLoS One 2014; 9:e93060. [PMID: 24755653 PMCID: PMC3995651 DOI: 10.1371/journal.pone.0093060] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 03/02/2014] [Indexed: 11/18/2022] Open
Abstract
The regulation of membrane shapes is central to many cellular phenomena. Bin/Amphiphysin/Rvs (BAR) domain-containing proteins are key players for membrane remodeling during endocytosis, cell migration, and endosomal sorting. BIN1, which contains an N-BAR domain, is assumed to be essential for biogenesis of plasma membrane invaginations (T-tubules) in muscle tissues. Three mutations, K35N, D151N and R154Q, have been discovered so far in the BAR domain of BIN1 in patients with centronuclear myopathy (CNM), where impaired organization of T-tubules has been reported. However, molecular mechanisms behind this malfunction have remained elusive. None of the BIN1 disease mutants displayed a significantly compromised curvature sensing ability. However, two mutants showed impaired membrane tubulation both in vivo and in vitro, and displayed characteristically different behaviors. R154Q generated smaller membrane curvature compared to WT N-BAR. Quantification of protein density on membranes revealed a lower membrane-bound density for R154Q compared to WT and the other mutants, which appeared to be the primary reason for the observation of impaired deformation capacity. The D151N mutant was unable to tubulate liposomes under certain experimental conditions. At medium protein concentrations we found 'budding' structures on liposomes that we hypothesized to be intermediates during the tubulation process except for the D151N mutant. Chemical crosslinking assays suggested that the D151N mutation impaired protein oligomerization upon membrane binding. Although we found an insignificant difference between WT and K35N N-BAR in in vitro assays, depolymerizing actin in live cells allowed tubulation of plasma membranes through the K35N mutant. Our results provide insights into the membrane-involved pathophysiological mechanisms leading to human disease.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Zheng Shi
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tobias Baumgart
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
24
|
Prendergast GC, Smith C, Thomas S, Mandik-Nayak L, Laury-Kleintop L, Metz R, Muller AJ. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother 2014; 63:721-35. [PMID: 24711084 DOI: 10.1007/s00262-014-1549-4] [Citation(s) in RCA: 390] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/26/2014] [Indexed: 12/15/2022]
Abstract
Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-β signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.
Collapse
Affiliation(s)
- George C Prendergast
- Lankenau Institute for Medical Research (LIMR), 100 Lancaster Avenue, Wynnewood, PA, 19096, USA,
| | | | | | | | | | | | | |
Collapse
|
25
|
Simionescu-Bankston A, Leoni G, Wang Y, Pham PP, Ramalingam A, DuHadaway JB, Faundez V, Nusrat A, Prendergast GC, Pavlath GK. The N-BAR domain protein, Bin3, regulates Rac1- and Cdc42-dependent processes in myogenesis. Dev Biol 2013; 382:160-71. [PMID: 23872330 DOI: 10.1016/j.ydbio.2013.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/01/2013] [Accepted: 07/05/2013] [Indexed: 11/28/2022]
Abstract
Actin dynamics are necessary at multiple steps in the formation of multinucleated muscle cells. BAR domain proteins can regulate actin dynamics in several cell types, but have been little studied in skeletal muscle. Here, we identify novel functions for the N-BAR domain protein, Bridging integrator 3 (Bin3), during myogenesis in mice. Bin3 plays an important role in regulating myofiber size in vitro and in vivo. During early myogenesis, Bin3 promotes migration of differentiated muscle cells, where it colocalizes with F-actin in lamellipodia. In addition, Bin3 forms a complex with Rac1 and Cdc42, Rho GTPases involved in actin polymerization, which are known to be essential for myotube formation. Importantly, a Bin3-dependent pathway is a major regulator of Rac1 and Cdc42 activity in differentiated muscle cells. Overall, these data classify N-BAR domain proteins as novel regulators of actin-dependent processes in myogenesis, and further implicate BAR domain proteins in muscle growth and repair.
Collapse
Affiliation(s)
- Adriana Simionescu-Bankston
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Amphiphysin I but not dynamin I nor synaptojanin mRNA expression increased after repeated methamphetamine administration in the rat cerebrum and cerebellum. J Neural Transm (Vienna) 2012; 120:1039-52. [PMID: 23224692 DOI: 10.1007/s00702-012-0931-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 11/16/2012] [Indexed: 01/13/2023]
Abstract
Dopamine increases/decreases synaptic vesicle recycling and in schizophrenia the proteins/mRNA is decreased. We isolated cDNA clone, similar to amphiphysin 1 (vesicle protein) mRNA from the neocortex of rats injected repeatedly with methamphetamine using polymerase chain reaction (PCR) differential display. This clone is highly homologous to the 3' region of the human amphiphysin gene. PCR extension study using a primer specific for the rat amphiphysin 1 gene and a primer located within the clone revealed that it is the 3' UTR region of the rat amphiphysin 1 gene. Furthermore, in situ hybridization revealed that amphiphysin 1 mRNA is expressed in the cerebrum, medial thalamus, hippocampus and cerebellum. In the cerebellum, amphiphysin mRNA expression was confined to upper granule cell layer. Repeated methamphetamine administration increased amphiphysin I mRNA expression in both anterior part of the cerebrum, and the cerebellum. However, the repeated administration did not alter mRNA expression of the other vesicle proteins, synaptotagmin I, synapsin I, synaptojanin and dynamin I, we conclude that the repeated administration selectively increased amphiphysin 1 mRNA expression. Thus, amphiphysin 1 does not work as synaptic recycling, but it is suggested, as a part of pathogenesis of brain tissue injury (under Ca²⁺ and Mg²⁺ devoid environment) in repeated methamphetamine-injected states, the gene regulate actin-asssembly, learning, cell stress signaling and cell polarity.
Collapse
|
27
|
Abstract
Stimulation of the receptor tyrosine kinase KIT by Stem Cell Factor (SCF) triggers activation of RAS and its downstream effectors. Proper KIT activation is essential for the maturation, survival and proliferation of mast cells. In addition, SCF activation of KIT is critical for recruiting mast cells to sites of infection or injury, where they release a mix of pro-inflammatory substances. RIN3, a RAS effector and RAB5-directed guanine nucleotide exchange factor (GEF), is highly expressed and enriched in human mast cells. SCF treatment of mast cells increased the amount of GTP-bound RAB5, and the degree of RAB5 activation correlated with the expression level of RIN3. At the same time, SCF caused the dissociation of a pre-formed complex of RIN3 with BIN2, a membrane bending protein implicated in endocytosis. Silencing of RIN3 increased the rate of SCF-induced KIT internalization, while persistent RIN3 over-expression led to KIT down regulation. These observations strongly support a role for RIN3 in coordinating the early steps of KIT endocytosis. Importantly, RIN3 also functioned as an inhibitor of mast cell migration toward SCF. Finally, we demonstrate that elevated RIN3 levels sensitize mastocytosis cells to treatment with a KIT tyrosine kinase inhibitor, suggesting the value of a two-pronged inhibitor approach for this difficult to treat malignancy. These findings directly connect KIT activation with a mast cell-specific RAS effector that regulates the cellular response to SCF and provide new insight for the development of more effective mastocytosis treatments.
Collapse
Affiliation(s)
- Christine Janson
- Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Noriyuki Kasahara
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United of States of America
| | - John Colicelli
- Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Schellenberg GD, Montine TJ. The genetics and neuropathology of Alzheimer's disease. Acta Neuropathol 2012; 124:305-23. [PMID: 22618995 DOI: 10.1007/s00401-012-0996-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 02/07/2023]
Abstract
Here we review the genetic causes and risks for Alzheimer's disease (AD). Early work identified mutations in three genes that cause AD: APP, PSEN1 and PSEN2. Although mutations in these genes are rare causes of AD, their discovery had a major impact on our understanding of molecular mechanisms of AD. Early work also revealed the ε4 allele of the APOE as a strong risk factor for AD. Subsequently, SORL1 also was identified as an AD risk gene. More recently, advances in our knowledge of the human genome, made possible by technological advances and methods to analyze genomic data, permit systematic identification of genes that contribute to AD risk. This work, so far accomplished through single nucleotide polymorphism arrays, has revealed nine new genes implicated in AD risk (ABCA7, BIN1, CD33, CD2AP, CLU, CR1, EPHA1, MS4A4E/MS4A6A, and PICALM). We review the relationship between these mutations and genetic variants and the neuropathologic features of AD and related disorders. Together, these discoveries point toward a new era in neurodegenerative disease research that impacts not only AD but also related illnesses that produce cognitive and behavioral deficits.
Collapse
Affiliation(s)
- Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6100, USA.
| | | |
Collapse
|
29
|
Kelsey JS, Fastman NM, Noratel EF, Blumberg DD. Ndm, a coiled-coil domain protein that suppresses macropinocytosis and has effects on cell migration. Mol Biol Cell 2012; 23:3407-19. [PMID: 22809629 PMCID: PMC3431939 DOI: 10.1091/mbc.e12-05-0392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The ampA gene has a role in cell migration in Dictyostelium discoideum. Cells overexpressing AmpA show an increase in cell migration, forming large plaques on bacterial lawns. A second-site suppressor of this ampA-overexpressing phenotype identified a previously uncharacterized gene, ndm, which is described here. The Ndm protein is predicted to contain a coiled-coil BAR-like domain-a domain involved in endocytosis and membrane bending. ndm-knockout and Ndm-monomeric red fluorescent protein-expressing cell lines were used to establish a role for ndm in suppressing endocytosis. An increase in the rate of endocytosis and in the number of endosomes was detected in ndm(-) cells. During migration ndm(-) cells formed numerous endocytic cups instead of the broad lamellipodia structure characteristic of moving cells. A second lamellipodia-based function-cell spreading-was also defective in the ndm(-) cells. The increase in endocytosis and the defect in lamellipodia formation were associated with reduced chemotaxis in ndm(-) cells. Immunofluorescence results and glutathione S-transferase pull-down assays revealed an association of Ndm with coronin and F-actin. The results establish ndm as a gene important in regulating the balance between formation of endocytic cups and lamellipodia structures.
Collapse
Affiliation(s)
- Jessica S Kelsey
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | | | | | | |
Collapse
|
30
|
Chang MY, Boulden J, Valenzano MC, Soler AP, Muller AJ, Mullin JM, Prendergast GC. Bin1 attenuation suppresses experimental colitis by enforcing intestinal barrier function. Dig Dis Sci 2012; 57:1813-21. [PMID: 22526583 PMCID: PMC3677578 DOI: 10.1007/s10620-012-2147-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 03/16/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is associated with defects in intestinal barriers that rely upon cellular tight junctions. Thus, identifying genes that could be targeted to enforce tight junctions and improve barrier function may lead to new treatment strategies for IBD. AIMS This preclinical study aimed to evaluate an hypothesized role for the tumor suppressor gene Bin1 as a modifier of the severity of experimental colitis. METHODS We ablated the Bin1 gene in a mosaic mouse model to evaluate its effects on experimental colitis and intestinal barrier function. Gross pathology, histology and inflammatory cytokine expression patterns were characterized and ex vivo physiology determinations were conducted to evaluate barrier function in intact colon tissue. RESULTS Bin1 attenuation limited experimental colitis in a sexually dimorphic manner with stronger protection in female subjects. Colitis suppression was associated with an increase in basal transepithelial electrical resistance (TER) and a decrease in paracellular transepithelial flux, compared to control wild-type animals. In contrast, Bin1 attenuation did not affect short circuit current, nor did it alter the epithelial barrier response to non-inflammatory permeability enhancers in the absence of inflammatory stimuli. CONCLUSIONS Bin1 is a genetic modifier of experimental colitis that controls the paracellular pathway of transcellular ion transport regulated by cellular tight junctions. Our findings offer a preclinical validation of Bin1 as a novel therapeutic target for IBD treatment.
Collapse
Affiliation(s)
| | | | | | - Alejandro P. Soler
- Lankenau Institute for Medical Research, Wynnewood PA USA,Richfield Laboratory of Dermatopathology, Cincinnati OH USA
| | | | | | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood PA USA,Department of Pathology, Anatomy & Cell Biology, Jefferson Medical College and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA USA
| |
Collapse
|
31
|
Esmailzadeh S, Jiang X. AHI-1: a novel signaling protein and potential therapeutic target in human leukemia and brain disorders. Oncotarget 2012; 2:918-34. [PMID: 22248740 PMCID: PMC3282096 DOI: 10.18632/oncotarget.405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Progress in the understanding of the molecular and cellular mechanisms of human cancer, including human leukemia and lymphomas, has been spurred by cloning of fusion genes created by chromosomal translocations or by retroviral insertional mutagenesis; a number of oncogenes and tumor suppressors involved in development of a number of malignancies have been identified in this manner. The BCR-ABL fusion gene, originating in a multipotent hematopoietic stem cell, is the molecular signature of chronic myeloid leukemia (CML). Discovery of this fusion gene has led to the development of one of the first successful targeted molecular therapies for cancer (Imatinib). It illustrates the advances that can result from an understanding of the molecular basis of disease. However, there still remain many as yet unidentified mutations that may influence the initiation or progression of human diseases. Thus, identification and characterization of the mechanism of action of genes that contribute to human diseases is an important and opportune area of current research. One promising candidate as a potential therapeutic target is Abelson helper integration site-1(Ahi-1/AHI-1) that was identified by retroviral insertional mutagenesis in murine models of leukemia/lymphomas and is highly elevated in certain human lymphoma and leukemia stem/progenitor cells. It encodes a unique protein with a SH3 domain, multiple SH3 binding sites and a WD40-repeat domain, suggesting that the normal protein has novel signaling activities. A new AHI-1-BCR-ABL-JAK2 interaction complex has recently been identified and this complex regulates transforming activities and drug resistance in CML stem/progenitor cells. Importantly, AHI-1 has recently been identified as a susceptibility gene involved in a number of brain disorders, including Joubert syndrome. Therefore, understanding molecular functions of the AHI-1 gene could lead to important and novel insights into disease processes involved in specific types of diseases. Ultimately, this knowledge will set the stage for translation into new and more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Sharmin Esmailzadeh
- Terry Fox Laboratory, British Columbia Cancer Agency and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
32
|
Chen Y, Aardema J, Misra A, Corey SJ. BAR proteins in cancer and blood disorders. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2012; 3:198-208. [PMID: 22773959 PMCID: PMC3388730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 04/18/2012] [Indexed: 06/01/2023]
Abstract
Remodeling of the membrane and cytoskeleton is involved in a wide range of normal and pathologic cellular function. These are complex, highly-coordinated biochemical and biophysical processes involving dozens of proteins. Serving as a scaffold for a variety of proteins and possessing a domain that interacts with plasma membranes, the BAR family of proteins contribute to a range of cellular functions characterized by membrane and cytoskeletal remodeling. There are several subgroups of BAR proteins: BAR, N-BAR, I-BAR, and F-BAR. They differ in their ability to induce angles of membrane curvature and in their recruitment of effector proteins. Evidence is accumulating that BAR proteins contribute to cancer cell invasion, T cell trafficking, phagocytosis, and platelet production. In this review, we discuss the physiological function of BAR proteins and discuss how they contribute to blood and cancer disorders.
Collapse
Affiliation(s)
- Yolande Chen
- Departments of Pediatrics and Cell & Molecular Biology, Children’s Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of MedicineChicago, IL
| | - Jorie Aardema
- Departments of Pediatrics and Cell & Molecular Biology, Children’s Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of MedicineChicago, IL
| | - Ashish Misra
- Division of Cardiology, Department of Medicine, Yale University School of MedicineNew Haven, CT, USA
| | - Seth J Corey
- Departments of Pediatrics and Cell & Molecular Biology, Children’s Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of MedicineChicago, IL
| |
Collapse
|
33
|
McKenna ES, Tamayo P, Cho YJ, Tillman EJ, Mora-Blanco EL, Sansam CG, Koellhoffer EC, Pomeroy SL, Roberts CWM. Epigenetic inactivation of the tumor suppressor BIN1 drives proliferation of SNF5-deficient tumors. Cell Cycle 2012; 11:1956-65. [PMID: 22544318 DOI: 10.4161/cc.20280] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Emerging evidence demonstrates that subunits of the SWI/SNF chromatin remodeling complex are specifically mutated at high frequency in a variety of human cancer types. SNF5 (SMARCB1/INI1/BAF47), a core subunit of the SWI/SNF complex, is inactivated in the vast majority of rhabdoid tumors (RT), an aggressive type of pediatric cancer. SNF5-deficient cancers are diploid and genomically stable, suggesting that epigenetically based changes in transcription are key drivers of tumor formation caused by SNF5 loss. However, there is limited understanding of the target genes that drive cancer formation following SNF5 loss. Here we performed comparative expression analyses upon three independent SNF5-deficient cancer data sets from both human and mouse and identify downregulation of the BIN1 tumor suppressor as a conserved event in primary SNF5-deficient cancers. We show that SNF5 recruits the SWI/SNF complex to the BIN1 promoter, and that the marked reduction of BIN1 expression in RT correlates with decreased SWI/SNF occupancy. Functionally, we demonstrate that re-expression of BIN1 specifically compromises the proliferation of SNF5-deficient RT cell lines. Identification of BIN1 as a SNF5 target gene reveals a novel tumor suppressive regulatory mechanism whose disruption can drive cancer formation.
Collapse
Affiliation(s)
- Elizabeth S McKenna
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lundgaard GL, Daniels NE, Pyndiah S, Cassimere EK, Ahmed KM, Rodrigue A, Kihara D, Post CB, Sakamuro D. Identification of a novel effector domain of BIN1 for cancer suppression. J Cell Biochem 2012; 112:2992-3001. [PMID: 21678469 DOI: 10.1002/jcb.23222] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bridging integrator 1 (BIN1) is a nucleocytoplasmic adaptor protein with tumor suppressor properties. The protein interacts with and inhibits the c-MYC transcription factor through the BIN1 MYC-binding domain (MBD). However, in vitro colony formation assays have clearly demonstrated that the MBD is not essential for BIN1-mediated growth arrest. We hypothesized that BIN1 contains a MYC-independent effector domain (MID) for cancer suppression. Because a functionally unique domain frequently contains a distinct structure, the human full-length BIN1 protein was subjected to limited trypsin digestion and the digested peptides were analyzed with Edman sequencing and mass spectrometry. We identified a trypsin-resistant peptide that corresponds to amino acids 146-268 of BIN1. It encompassed part of the BAR region, a putative effector region of BIN1. Computational analysis predicted that the peptide is very likely to exhibit coiled-coil motifs, implying a potential role for this region in sustaining the BIN1 structure and function. Like MBD-deleted BIN1, the trypsin-resistant peptide of BIN1 was predominantly present in the cytoplasm and was sufficient to inhibit cancer growth, regardless of dysregulated c-MYC activity. Our results suggest that the coiled-coil BIN1 BAR peptide encodes a novel BIN1 MID domain, through which BIN1 acts as a MYC-independent cancer suppressor.
Collapse
Affiliation(s)
- Greta L Lundgaard
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Neuvonen M, Kazlauskas A, Martikainen M, Hinkkanen A, Ahola T, Saksela K. SH3 domain-mediated recruitment of host cell amphiphysins by alphavirus nsP3 promotes viral RNA replication. PLoS Pathog 2011; 7:e1002383. [PMID: 22114558 PMCID: PMC3219718 DOI: 10.1371/journal.ppat.1002383] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 10/02/2011] [Indexed: 01/08/2023] Open
Abstract
Among the four non-structural proteins of alphaviruses the function of nsP3 is the least well understood. NsP3 is a component of the viral replication complex, and composed of a conserved aminoterminal macro domain implicated in viral RNA synthesis, and a poorly conserved carboxyterminal region. Despite the lack of overall homology we noted a carboxyterminal proline-rich sequence motif shared by many alphaviral nsP3 proteins, and found it to serve as a preferred target site for the Src-homology 3 (SH3) domains of amphiphysin-1 and -2. Nsp3 proteins of Semliki Forest (SFV), Sindbis (SINV), and Chikungunya viruses all showed avid and SH3-dependent binding to amphiphysins. Upon alphavirus infection the intracellular distribution of amphiphysin was dramatically altered and colocalized with nsP3. Mutations in nsP3 disrupting the amphiphysin SH3 binding motif as well as RNAi-mediated silencing of amphiphysin-2 expression resulted in impaired viral RNA replication in HeLa cells infected with SINV or SFV. Infection of Balb/c mice with SFV carrying an SH3 binding-defective nsP3 was associated with significantly decreased mortality. These data establish SH3 domain-mediated binding of nsP3 with amphiphysin as an important host cell interaction promoting alphavirus replication. The genus Alphavirus contains 29 known species that are transmitted by arthropods and include many important pathogens, such as Chikungunya virus (CHKV), which during the past decade has re-emerged to cause massive epidemics of febrile arthralgia around the Indian Ocean. The role of the alphaviral non-structural protein 3 (nsP3) has been linked to RNA replication and disease pathogenesis, but its molecular functions have remained elusive. Here we show that the nsP3s of CHKV as well as Sindbis and Semliki Forest viruses use a conserved proline-rich motif to interact with the Src-homology-3 (SH3) domain of host cell amphiphysins Amph1 and BIN1/Amph2, two adaptor proteins prominently involved in cellular membrane dynamics. We observed a striking re-localization of amphiphysin to alphaviral replication complexes in infected cells, and found that disruption of the amphiphysin SH3 binding motif in nsP3 strongly suppressed virus replication in vitro and attenuated Semliki Forest virus in infected mice. Thus, we conclude that amphiphysins are novel and important host cell factors involved in supporting alphavirus replication.
Collapse
Affiliation(s)
- Maarit Neuvonen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Arunas Kazlauskas
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Miika Martikainen
- A. I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland and the Cancer Center of Eastern Finland, Kuopio, Finland
| | - Ari Hinkkanen
- A. I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland and the Cancer Center of Eastern Finland, Kuopio, Finland
| | - Tero Ahola
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kalle Saksela
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- * E-mail:
| |
Collapse
|
36
|
Abstract
The MTM (myotubularin)/MTMR (myotubularin-related) protein family is comprised of 15 lipid phosphatases, of which nine members are catalytically active. MTMs are known to play a fundamental role in human physiology as gene mutations can give rise to X-linked myotubular myopathy or Charcot-Marie-Tooth disease, which manifest in skeletal muscle or in peripheral neurons respectively. Interestingly, studies have shown MTMR2 and MTMR5, two MTM family members, to be highly expressed in the testis, particularly in Sertoli and germ cells, and knockout of either gene resulted in spermatogenic defects. Other studies have shown that MTMR2 functions in endocytosis and membrane trafficking. In the testis, MTMR2 interacts and co-localizes with c-Src/phospho-Src-(Tyr⁴¹⁶), a non-receptor protein tyrosine kinase that regulates the phosphorylation state of proteins at the apical ES (ectoplasmic specialization), a unique type of cell junction found between Sertoli cells and elongating/elongated spermatids. In the present review, we highlight recent findings that have made a significant impact on our understanding of this protein family in normal cell function and in disease, with the emphasis on the role of MTMs and MTMRs in spermatogenesis. We also describe a working model to explain how MTMR2 interacts with other proteins such as c-Src, dynamin 2, EPS8 (growth factor receptor pathway substrate 8) and ARP2/3 (actin-related protein 2/3) at the apical ES and the apical TBC (tubulobulbar complex; tubular-like invaginations that function in the disassembly of the apical ES and in the recycling of its components) to regulate spermiation at late stage VIII of the seminiferous epithelial cycle.
Collapse
|
37
|
Liao XH, Buggey J, Kimmel AR. Chemotactic activation of Dictyostelium AGC-family kinases AKT and PKBR1 requires separate but coordinated functions of PDK1 and TORC2. J Cell Sci 2010; 123:983-92. [PMID: 20200230 DOI: 10.1242/jcs.064022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Protein kinases AKT and PKBR1 of Dictyostelium belong to the AGC protein kinase superfamily. AKT and PKBR1 are phosphorylated at similar sites by phosphoinositide-dependent kinase 1 (PDK1) and TORC2 kinases; however, they have different subcellular localizing domains. AKT has a phosphoinositide 3-kinase (PI3K)/phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)]-regulated PH (pleckstrin homology) domain whereas PKBR1 is myristoylated and persistently membrane localized. Using strains defective for PI3K/PtdIns(3,4,5)P(3)-, PDK1- and TORC2-signaling or strains that express phospho-site mutants of AKT and PKBR1, we dissect the different roles of PI3K/PtdIns(3,4,5)P(3), PDK1 and TORC2. We show that activation of AKT and PKBR1 requires PDK1-site phosphorylation, but that phosphorylation by TORC2 is insufficient for AKT or PKBR1 activation. However, PDK1-site phosphorylation is dependent on phosphorylation by TORC2, which suggests that there is regulatory coordination among PDK1, TORC2 and their phospho-site targets. This defines a separate input for signaling in control of chemotaxis and dependency on PDK1 function. We also demonstrate that PDK1 in Dictyostelium functions independently of PI3K/PtdIns(3,4,5)P(3). Finally, we show that AKT and PKBR1 exhibit substrate selectivity and identify two novel lipid-interacting proteins preferentially phosphorylated by AKT. Despite certain similarities, AKT and PKBR1 have distinct regulatory paths that impact activation and effector targeting, with PDK1 serving a central role.
Collapse
Affiliation(s)
- Xin-Hua Liao
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892-8028, USA
| | | | | |
Collapse
|
38
|
Prendergast GC, Metz R, Muller AJ. Towards a genetic definition of cancer-associated inflammation: role of the IDO pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2082-7. [PMID: 20228228 DOI: 10.2353/ajpath.2010.091173] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chronic inflammation drives the development of many cancers, but a genetic definition of what constitutes 'cancer-associated' inflammation has not been determined. Recently, a mouse genetic study revealed a critical role for the immune escape mediator indoleamine 2,3-dioxygenase (IDO) in supporting inflammatory skin carcinogenesis. IDO is generally regarded as being immunosuppressive; however, there was no discernable difference in generalized inflammatory processes in IDO-null mice under conditions where tumor development was significantly suppressed, implicating IDO as key to establishing the pathogenic state of 'cancer-associated' inflammation. Here we review recent findings and their potential implications to understanding the relationship between immune escape and inflammation in cancer. Briefly, we propose that genetic pathways of immune escape in cancer are synonymous with pathways that define 'cancer-associated' inflammation and that these processes may be identical rather than distinct, as generally presumed, in terms of their genetic definition.
Collapse
|
39
|
From dynamic live cell imaging to 3D ultrastructure: novel integrated methods for high pressure freezing and correlative light-electron microscopy. PLoS One 2010; 5:e9014. [PMID: 20140253 PMCID: PMC2815783 DOI: 10.1371/journal.pone.0009014] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 01/11/2010] [Indexed: 11/19/2022] Open
Abstract
Background In cell biology, the study of proteins and organelles requires the combination of different imaging approaches, from live recordings with light microscopy (LM) to electron microscopy (EM). Methodology To correlate dynamic events in adherent cells with both ultrastructural and 3D information, we developed a method for cultured cells that combines confocal time-lapse images of GFP-tagged proteins with electron microscopy. With laser micro-patterned culture substrate, we created coordinates that were conserved at every step of the sample preparation and visualization processes. Specifically designed for cryo-fixation, this method allowed a fast freezing of dynamic events within seconds and their ultrastructural characterization. We provide examples of the dynamic oligomerization of GFP-tagged myotubularin (MTM1) phosphoinositides phosphatase induced by osmotic stress, and of the ultrastructure of membrane tubules dependent on amphiphysin 2 (BIN1) expression. Conclusion Accessible and versatile, we show that this approach is efficient to routinely correlate functional and dynamic LM with high resolution morphology by EM, with immuno-EM labeling, with 3D reconstruction using serial immuno-EM or tomography, and with scanning-EM.
Collapse
|
40
|
Pyrzynska B, Pilecka I, Miaczynska M. Endocytic proteins in the regulation of nuclear signaling, transcription and tumorigenesis. Mol Oncol 2009; 3:321-38. [PMID: 19577966 DOI: 10.1016/j.molonc.2009.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 06/01/2009] [Accepted: 06/02/2009] [Indexed: 01/14/2023] Open
Abstract
Accumulating evidence argues that many proteins governing membrane sorting during endocytosis participate also in nuclear signaling and transcriptional regulation, mostly by modulating the activity of various nuclear factors. Some adaptors and accessory proteins acting in clathrin-mediated internalization, as well as endosomal sorting proteins can undergo nuclear translocation and affect gene expression directly, while for others the effects may be more indirect. Although it is often unclear to what extent the endocytic and nuclear functions are interrelated, several of such proteins are implicated in the regulation of cell proliferation and tumorigenesis, arguing that their dual-function nature may be of physiological importance.
Collapse
Affiliation(s)
- Beata Pyrzynska
- International Institute of Molecular and Cell Biology, Laboratory of Cell Biology, 02-109 Warsaw, Poland
| | | | | |
Collapse
|
41
|
Caino MC, Meshki J, Kazanietz MG. Hallmarks for senescence in carcinogenesis: novel signaling players. Apoptosis 2009; 14:392-408. [PMID: 19169823 DOI: 10.1007/s10495-009-0316-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|