1
|
Arms LM, Duchatel RJ, Jackson ER, Sobrinho PG, Dun MD, Hua S. Current status and advances to improving drug delivery in diffuse intrinsic pontine glioma. J Control Release 2024; 370:835-865. [PMID: 38744345 DOI: 10.1016/j.jconrel.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Diffuse midline glioma (DMG), including tumors diagnosed in the brainstem (diffuse intrinsic pontine glioma - DIPG), is the primary cause of brain tumor-related death in pediatric patients. DIPG is characterized by a median survival of <12 months from diagnosis, harboring the worst 5-year survival rate of any cancer. Corticosteroids and radiation are the mainstay of therapy; however, they only provide transient relief from the devastating neurological symptoms. Numerous therapies have been investigated for DIPG, but the majority have been unsuccessful in demonstrating a survival benefit beyond radiation alone. Although many barriers hinder brain drug delivery in DIPG, one of the most significant challenges is the blood-brain barrier (BBB). Therapeutic compounds must possess specific properties to enable efficient passage across the BBB. In brain cancer, the BBB is referred to as the blood-brain tumor barrier (BBTB), where tumors disrupt the structure and function of the BBB, which may provide opportunities for drug delivery. However, the biological characteristics of the brainstem's BBB/BBTB, both under normal physiological conditions and in response to DIPG, are poorly understood, which further complicates treatment. Better characterization of the changes that occur in the BBB/BBTB of DIPG patients is essential, as this informs future treatment strategies. Many novel drug delivery technologies have been investigated to bypass or disrupt the BBB/BBTB, including convection enhanced delivery, focused ultrasound, nanoparticle-mediated delivery, and intranasal delivery, all of which are yet to be clinically established for the treatment of DIPG. Herein, we review what is known about the BBB/BBTB and discuss the current status, limitations, and advances of conventional and novel treatments to improving brain drug delivery in DIPG.
Collapse
Affiliation(s)
- Lauren M Arms
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Ryan J Duchatel
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Evangeline R Jackson
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Pedro Garcia Sobrinho
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Matthew D Dun
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
2
|
Saratsis AM, Knowles T, Petrovic A, Nazarian J. H3K27M mutant glioma: Disease definition and biological underpinnings. Neuro Oncol 2024; 26:S92-S100. [PMID: 37818718 PMCID: PMC11066930 DOI: 10.1093/neuonc/noad164] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Indexed: 10/12/2023] Open
Abstract
High-grade glioma (HGG) is the most common cause of cancer death in children and the most common primary central nervous system tumor in adults. While pediatric HGG was once thought to be biologically similar to the adult form of disease, research has shown these malignancies to be significantly molecularly distinct, necessitating distinct approaches to their clinical management. However, emerging data have shown shared molecular events in pediatric and adult HGG including the histone H3K27M mutation. This somatic missense mutation occurs in genes encoding one of two isoforms of the Histone H3 protein, H3F3A (H3.3), or HIST1H3B (H3.1), and is detected in up to 80% of pediatric diffuse midline gliomas and in up to 60% of adult diffuse gliomas. Importantly, the H3K27M mutation is associated with poorer overall survival and response to therapy compared to patients with H3 wild-type tumors. Here, we review the clinical features and biological underpinnings of pediatric and adult H3K27M mutant glioma, offering a groundwork for understanding current research and clinical approaches for the care of patients suffering with this challenging disease.
Collapse
Affiliation(s)
| | | | - Antonela Petrovic
- DMG Research Center, Department of Oncology, University Children’s Hospital, University of Zürich, Zürich, Switzerland
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children’s National Health System, Washington, District of Columbia, USA
- Brain Tumor Institute, Children’s National Health System, Washington, District of Columbia, USA
- DMG Research Center, Department of Pediatrics, University Children’s Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
3
|
Ocasio JK, Budd KM, Roach JT, Andrews JM, Baker SJ. Oncohistones and disrupted development in pediatric-type diffuse high-grade glioma. Cancer Metastasis Rev 2023; 42:367-388. [PMID: 37119408 PMCID: PMC10441521 DOI: 10.1007/s10555-023-10105-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/05/2023] [Indexed: 05/01/2023]
Abstract
Recurrent, clonal somatic mutations in histone H3 are molecular hallmarks that distinguish the genetic mechanisms underlying pediatric and adult high-grade glioma (HGG), define biological subgroups of diffuse glioma, and highlight connections between cancer, development, and epigenetics. These oncogenic mutations in histones, now termed "oncohistones", were discovered through genome-wide sequencing of pediatric diffuse high-grade glioma. Up to 80% of diffuse midline glioma (DMG), including diffuse intrinsic pontine glioma (DIPG) and diffuse glioma arising in other midline structures including thalamus or spinal cord, contain histone H3 lysine 27 to methionine (K27M) mutations or, rarely, other alterations that result in a depletion of H3K27me3 similar to that induced by H3 K27M. This subgroup of glioma is now defined as diffuse midline glioma, H3K27-altered. In contrast, histone H3 Gly34Arg/Val (G34R/V) mutations are found in approximately 30% of diffuse glioma arising in the cerebral hemispheres of older adolescents and young adults, now classified as diffuse hemispheric glioma, H3G34-mutant. Here, we review how oncohistones modulate the epigenome and discuss the mutational landscape and invasive properties of histone mutant HGGs of childhood. The distinct mechanisms through which oncohistones and other mutations rewrite the epigenetic landscape provide novel insights into development and tumorigenesis and may present unique vulnerabilities for pHGGs. Lessons learned from these rare incurable brain tumors of childhood may have broader implications for cancer, as additional high- and low-frequency oncohistone mutations have been identified in other tumor types.
Collapse
Affiliation(s)
- Jennifer K Ocasio
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kaitlin M Budd
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, USA
| | - Jordan T Roach
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, USA
- College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Jared M Andrews
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, USA.
| |
Collapse
|
4
|
K27M in canonical and noncanonical H3 variants occurs in distinct oligodendroglial cell lineages in brain midline gliomas. Nat Genet 2022; 54:1865-1880. [PMID: 36471070 PMCID: PMC9742294 DOI: 10.1038/s41588-022-01205-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/16/2022] [Indexed: 12/12/2022]
Abstract
Canonical (H3.1/H3.2) and noncanonical (H3.3) histone 3 K27M-mutant gliomas have unique spatiotemporal distributions, partner alterations and molecular profiles. The contribution of the cell of origin to these differences has been challenging to uncouple from the oncogenic reprogramming induced by the mutation. Here, we perform an integrated analysis of 116 tumors, including single-cell transcriptome and chromatin accessibility, 3D chromatin architecture and epigenomic profiles, and show that K27M-mutant gliomas faithfully maintain chromatin configuration at developmental genes consistent with anatomically distinct oligodendrocyte precursor cells (OPCs). H3.3K27M thalamic gliomas map to prosomere 2-derived lineages. In turn, H3.1K27M ACVR1-mutant pontine gliomas uniformly mirror early ventral NKX6-1+/SHH-dependent brainstem OPCs, whereas H3.3K27M gliomas frequently resemble dorsal PAX3+/BMP-dependent progenitors. Our data suggest a context-specific vulnerability in H3.1K27M-mutant SHH-dependent ventral OPCs, which rely on acquisition of ACVR1 mutations to drive aberrant BMP signaling required for oncogenesis. The unifying action of K27M mutations is to restrict H3K27me3 at PRC2 landing sites, whereas other epigenetic changes are mainly contingent on the cell of origin chromatin state and cycling rate.
Collapse
|
5
|
Tomita Y, Shimazu Y, Somasundaram A, Tanaka Y, Takata N, Ishi Y, Gadd S, Hashizume R, Angione A, Pinero G, Hambardzumyan D, Brat DJ, Hoeman CM, Becher OJ. A novel mouse model of diffuse midline glioma initiated in neonatal oligodendrocyte progenitor cells highlights cell-of-origin dependent effects of H3K27M. Glia 2022; 70:1681-1698. [PMID: 35524725 PMCID: PMC9546478 DOI: 10.1002/glia.24189] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/13/2022]
Abstract
Diffuse midline glioma (DMG) is a type of lethal brain tumor that develops mainly in children. The majority of DMG harbor the K27M mutation in histone H3. Oligodendrocyte progenitor cells (OPCs) in the brainstem are candidate cells-of-origin for DMG, yet there is no genetically engineered mouse model of DMG initiated in OPCs. Here, we used the RCAS/Tv-a avian retroviral system to generate DMG in Olig2-expressing progenitors and Nestin-expressing progenitors in the neonatal mouse brainstem. PDGF-A or PDGF-B overexpression, along with p53 deletion, resulted in gliomas in both models. Exogenous overexpression of H3.3K27M had a significant effect on tumor latency and tumor cell proliferation when compared with H3.3WT in Nestin+ cells but not in Olig2+ cells. Further, the fraction of H3.3K27M-positive cells was significantly lower in DMGs initiated in Olig2+ cells relative to Nestin+ cells, both in PDGF-A and PDGF-B-driven models, suggesting that the requirement for H3.3K27M is reduced when tumorigenesis is initiated in Olig2+ cells. RNA-sequencing analysis revealed that the differentially expressed genes in H3.3K27M tumors were non-overlapping between Olig2;PDGF-B, Olig2;PDGF-A, and Nestin;PDGF-A models. GSEA analysis of PDGFA tumors confirmed that the transcriptomal effects of H3.3K27M are cell-of-origin dependent with H3.3K27M promoting epithelial-to-mesenchymal transition (EMT) and angiogenesis when Olig2 marks the cell-of-origin and inhibiting EMT and angiogenesis when Nestin marks the cell-of-origin. We did observe some overlap with H3.3K27M promoting negative enrichment of TNFA_Signaling_Via_NFKB in both models. Our study suggests that the tumorigenic effects of H3.3K27M are cell-of-origin dependent, with H3.3K27M being more oncogenic in Nestin+ cells than Olig2+ cells.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Department of Neurosurgery and Neuroendovascular SurgeryHiroshima City Hiroshima Citizens HospitalHiroshimaJapan
| | - Yosuke Shimazu
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Agila Somasundaram
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Yoshihiro Tanaka
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Arrhythmia Research, Department of CardiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Nozomu Takata
- Center for Vascular and Developmental BiologyFeinberg Cardiovascular and Renal Research Institute (FCVRRI), Northwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Yukitomo Ishi
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Samantha Gadd
- Department of PathologyAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Rintaro Hashizume
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Biochemistry and Molecular GeneticsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Angelo Angione
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Gonzalo Pinero
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Dolores Hambardzumyan
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Daniel J. Brat
- Department of PathologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Christine M. Hoeman
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Oren J. Becher
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Biochemistry and Molecular GeneticsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Jack Martin Division of Pediatric Hematology‐oncologyMount Sinai Kravis Children's HospitalNew YorkUSA
| |
Collapse
|
6
|
Cardona HJ, Somasundaram A, Crabtree DM, Gadd SL, Becher OJ. Prenatal overexpression of platelet-derived growth factor receptor A results in central nervous system hypomyelination. Brain Behav 2021; 11:e2332. [PMID: 34480532 PMCID: PMC8553322 DOI: 10.1002/brb3.2332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/01/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Platelet-derived growth factor (PDGF) signaling, through the ligand PDGF-A and its receptor PDGFRA, is important for the growth and maintenance of oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). PDGFRA signaling is downregulated prior to OPC differentiation into mature myelinating oligodendrocytes. By contrast, PDGFRA is often genetically amplified or mutated in many types of gliomas, including diffuse midline glioma (DMG) where OPCs are considered the most likely cell-of-origin. The cellular and molecular changes that occur in OPCs in response to unregulated PDGFRA expression, however, are not known. METHODS Here, we created a conditional knock-in (KI) mouse that overexpresses wild type (WT) human PDGFRA (hPDGFRA) in prenatal Olig2-expressing progenitors, and examined in vivo cellular and molecular consequences. RESULTS The KI mice exhibited stunted growth, ataxia, and a severe loss of myelination in the brain and spinal cord. When combined with the loss of p53, a tumor suppressor gene whose activity is decreased in DMG, the KI mice failed to develop tumors but still exhibited hypomyelination. RNA-sequencing analysis revealed decreased myelination gene signatures, indicating a defect in oligodendroglial development. Mice overexpressing PDGFRA in prenatal GFAP-expressing progenitors, which give rise to a broader lineage of cells than Olig2-progenitors, also developed myelination defects. CONCLUSION Our results suggest that embryonic overexpression of hPDGFRA in Olig2- or GFAP-progenitors is deleterious to OPC development and leads to CNS hypomyelination.
Collapse
Affiliation(s)
- Herminio Joey Cardona
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Agila Somasundaram
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Donna M Crabtree
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA.,Office of Clinical Research, Duke University Medical Center, Durham, NC, USA
| | - Samantha L Gadd
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Oren J Becher
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA.,Department of Pediatrics, Northwestern University, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
7
|
Vargas-Castro V, Gomez-Diaz R, Blanco-Alvarez VM, Tomas-Sanchez C, Gonzalez-Vazquez A, Aguilar-Peralta AK, Gonzalez-Barrios JA, Martinez-Fong D, Eguibar JR, Vivar C, Ugarte A, Soto-Rodriguez G, Brambila E, Millán-Perez-Peña L, Leon-Chavez BA. Long-term taurine administration improves motor skills in a tubulinopathy rat model by decreasing oxidative stress and promoting myelination. Mol Cell Neurosci 2021; 115:103643. [PMID: 34186187 DOI: 10.1016/j.mcn.2021.103643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 11/18/2022] Open
Abstract
The taiep rat undergoes hypomyelination and progressive demyelination caused by an abnormal microtubule accumulation in oligodendrocytes, which elicits neuroinflammation and motor behavior dysfunction. Based on taurine antioxidant and proliferative actions, this work explored whether its sustained administration from the embryonic age to adulthood could prevent neuroinflammation, stimulate cell proliferation, promote myelination, and relieve motor impairment. Taurine (50 mg/L of drinking water = 50 ppm) was given to taiep pregnant rats on gestational day 15 and afterward to the male offspring until eight months of age. We measured the levels of nitric oxide (NO), malondialdehyde + 4-hydroxyalkenals (MDA + 4-HDA), CXCL1, CXCR2 receptor, growth factors (BNDF and FGF2), cell proliferation, and myelin content over time. Integral motor behavior was also evaluated. Our results showed that taurine administration significantly decreased NO and MDA + 4-HDA levels, increased cell proliferation, and promoted myelination in an age- and brain region-dependent fashion compared with untreated taiep rats. Taurine effect on chemokines and growth factors was also variable. Taurine improved vestibular reflexes and limb muscular strength in perinatal rats and fine movements and immobility episodes in adult rats. These results show that chronic taurine administration partially alleviates the taiep neuropathology.
Collapse
Affiliation(s)
- Viridiana Vargas-Castro
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Ricardo Gomez-Diaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Victor M Blanco-Alvarez
- Facultad de Enfermería, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72304, Mexico
| | - Constantino Tomas-Sanchez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Alejandro Gonzalez-Vazquez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Ana Karina Aguilar-Peralta
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Juan A Gonzalez-Barrios
- Laboratorio de Medicina Genómica, Hospital Regional 1° de Octubre, ISSSTE, Mexico City C. P. 07760, Mexico
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, C. P. 07000 Mexico City, Mexico
| | - Jose R Eguibar
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72590, Mexico
| | - Carmen Vivar
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, C. P. 07000 Mexico City, Mexico
| | - Araceli Ugarte
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72590, Mexico
| | - Guadalupe Soto-Rodriguez
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72304, Mexico
| | - Eduardo Brambila
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Lourdes Millán-Perez-Peña
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico
| | - Bertha Alicia Leon-Chavez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue. C. P. 72570, Mexico.
| |
Collapse
|
8
|
Fauser M, Pan-Montojo F, Richter C, Kahle PJ, Schwarz SC, Schwarz J, Storch A, Hermann A. Chronic-Progressive Dopaminergic Deficiency Does Not Induce Midbrain Neurogenesis. Cells 2021; 10:775. [PMID: 33807497 PMCID: PMC8066763 DOI: 10.3390/cells10040775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Consecutive adult neurogenesis is a well-known phenomenon in the ventricular-subventricular zone of the lateral wall of the lateral ventricles (V-SVZ) and has been controversially discussed in so-called "non-neurogenic" brain areas such as the periventricular regions (PVRs) of the aqueduct and the fourth ventricle. Dopamine is a known modulator of adult neural stem cell (aNSC) proliferation and dopaminergic neurogenesis in the olfactory bulb, though a possible interplay between local dopaminergic neurodegeneration and induction of aNSC proliferation in mid/hindbrain PVRs is currently enigmatic. OBJECTIVE/HYPOTHESIS To analyze the influence of chronic-progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the V-SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of Parkinson´s disease (PD). METHODS We used Thy1-m[A30P]h α synuclein mice and Leu9'Ser hypersensitive α4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the V-SVZ, the aqueduct and the fourth ventricle. RESULTS In both animal models, overall proliferative activity in the V-SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the V-SVZ in Leu9'Ser hypersensitive α4* nAChR mice. Putative aNSCs in the mid/hindbrain PVRs are known to be quiescent in vivo in healthy controls, and dopaminergic deficiency did not induce proliferative activity in these regions in both disease models. CONCLUSIONS Our data do not support an activation of endogenous aNSCs in mid/hindbrain PVRs after local dopaminergic neurodegeneration. Spontaneous endogenous regeneration of dopaminergic cell loss through resident aNSCs is therefore unlikely.
Collapse
Affiliation(s)
- Mareike Fauser
- Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (M.F.); (A.S.)
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Francisco Pan-Montojo
- Munich Cluster for Systems Neurology, Department of Psychiatry, University Hospital LMU, 80336 Munich, Germany;
| | - Christian Richter
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Philipp J. Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, 72076 Tübingen, Germany;
- German Centre for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Sigrid C. Schwarz
- Department of Neurology, University Hospital Leipzig, 04103 Leipzig, Germany; (S.C.S.); (J.S.)
| | - Johannes Schwarz
- Department of Neurology, University Hospital Leipzig, 04103 Leipzig, Germany; (S.C.S.); (J.S.)
- Department of Neurology, Klinik Haag i. OB, 83527 Oberbayern, Germany
| | - Alexander Storch
- Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany; (M.F.); (A.S.)
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany;
- German Centre for Neurodegenerative Diseases (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Andreas Hermann
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany;
- German Centre for Neurodegenerative Diseases (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
| |
Collapse
|
9
|
Chen Z, Peng P, Zhang X, Mania-Farnell B, Xi G, Wan F. Advanced Pediatric Diffuse Pontine Glioma Murine Models Pave the Way towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13051114. [PMID: 33807733 PMCID: PMC7961799 DOI: 10.3390/cancers13051114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for ~15% of pediatric brain tumors, which invariably present with poor survival regardless of treatment mode. Several seminal studies have revealed that 80% of DIPGs harbor H3K27M mutation coded by HIST1H3B, HIST1H3C and H3F3A genes. The H3K27M mutation has broad effects on gene expression and is considered a tumor driver. Determination of the effects of H3K27M on posttranslational histone modifications and gene regulations in DIPG is critical for identifying effective therapeutic targets. Advanced animal models play critical roles in translating these cutting-edge findings into clinical trial development. Here, we review current molecular research progress associated with DIPG. We also summarize DIPG animal models, highlighting novel genomic engineered mouse models (GEMMs) and innovative humanized DIPG mouse models. These models will pave the way towards personalized precision medicine for the treatment of DIPGs.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Peng Peng
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Xiaolin Zhang
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Barbara Mania-Farnell
- Department of Biological Science, Purdue University Northwest, Hammond, IN 46323, USA;
| | - Guifa Xi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| | - Feng Wan
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| |
Collapse
|
10
|
Blackburn J, Chapur VF, Stephens JA, Zhao J, Shepler A, Pierson CR, Otero JJ. Revisiting the Neuropathology of Sudden Infant Death Syndrome (SIDS). Front Neurol 2020; 11:594550. [PMID: 33391159 PMCID: PMC7773837 DOI: 10.3389/fneur.2020.594550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/10/2020] [Indexed: 01/08/2023] Open
Abstract
Background: Sudden infant death syndrome (SIDS) is one of the leading causes of infant mortality in the United States (US). The extent to which SIDS manifests with an underlying neuropathological mechanism is highly controversial. SIDS correlates with markers of poor prenatal and postnatal care, generally rooted in the lack of access and quality of healthcare endemic to select racial and ethnic groups, and thus can be viewed in the context of health disparities. However, some evidence suggests that at least a subset of SIDS cases may result from a neuropathological mechanism. To explain these issues, a triple-risk hypothesis has been proposed, whereby an underlying biological abnormality in an infant facing an extrinsic risk during a critical developmental period SIDS is hypothesized to occur. Each SIDS decedent is thus thought to have a unique combination of these risk factors leading to their death. This article reviews the neuropathological literature of SIDS and uses machine learning tools to identify distinct subtypes of SIDS decedents based on epidemiological data. Methods: We analyzed US Period Linked Birth/Infant Mortality Files from 1990 to 2017 (excluding 1992–1994). Using t-SNE, an unsupervised machine learning dimensionality reduction algorithm, we identified clusters of SIDS decedents. Following identification of these groups, we identified changes in the rates of SIDS at the state level and across three countries. Results: Through t-SNE and distance based statistical analysis, we identified three groups of SIDS decedents, each with a unique peak age of death. Within the US, SIDS is geographically heterogeneous. Following this, we found low birth weight and normal birth weight SIDS rates have not been equally impacted by implementation of clinical guidelines. We show that across countries with different levels of cultural heterogeneity, reduction in SIDS rates has also been distinct between decedents with low vs. normal birth weight. Conclusions: Different epidemiological and extrinsic risk factors exist based on the three unique SIDS groups we identified with t-SNE and distance based statistical measurements. Clinical guidelines have not equally impacted the groups, and normal birth weight infants comprise more of the cases of SIDS even though low birth weight infants have a higher SIDS rate.
Collapse
Affiliation(s)
- Jessica Blackburn
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States.,Division of Anatomy, Department of Biomedical Education & Anatomy, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Valeria F Chapur
- Instituto de Ecoregiones Andinas (INECOA)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Biología de la Altura (INBIAL)/Universidad Nacional de Jujuy (UNJU), San Salvador de Jujuy, Argentina
| | - Julie A Stephens
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Jing Zhao
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Anne Shepler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States.,Franklin County Forensic Science Center, Columbus, OH, United States
| | - Christopher R Pierson
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States.,Division of Anatomy, Department of Biomedical Education & Anatomy, The Ohio State University College of Medicine, Columbus, OH, United States.,Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - José Javier Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
11
|
Weinstock NI, Kreher C, Favret J, Nguyen D, Bongarzone ER, Wrabetz L, Feltri ML, Shin D. Brainstem development requires galactosylceramidase and is critical for pathogenesis in a model of Krabbe disease. Nat Commun 2020; 11:5356. [PMID: 33097716 PMCID: PMC7584660 DOI: 10.1038/s41467-020-19179-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Krabbe disease (KD) is caused by a deficiency of galactosylceramidase (GALC), which induces demyelination and neurodegeneration due to accumulation of cytotoxic psychosine. Hematopoietic stem cell transplantation (HSCT) improves clinical outcomes in KD patients only if delivered pre-symptomatically. Here, we hypothesize that the restricted temporal efficacy of HSCT reflects a requirement for GALC in early brain development. Using a novel Galc floxed allele, we induce ubiquitous GALC ablation (Galc-iKO) at various postnatal timepoints and identify a critical period of vulnerability to GALC ablation between P4-6 in mice. Early Galc-iKO induction causes a worse KD phenotype, higher psychosine levels in the rodent brainstem and spinal cord, and a significantly shorter life-span of the mice. Intriguingly, GALC expression peaks during this critical developmental period in mice. Further analysis of this mouse model reveals a cell autonomous role for GALC in the development and maturation of immature T-box-brain-1 positive brainstem neurons. These data identify a perinatal developmental period, in which neuronal GALC expression influences brainstem development that is critical for KD pathogenesis.
Collapse
Affiliation(s)
- Nadav I Weinstock
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
| | - Conlan Kreher
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
| | - Jacob Favret
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
| | - Duc Nguyen
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA
| | - Daesung Shin
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA.
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA.
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA.
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), Buffalo, NY, 14214, USA.
| |
Collapse
|
12
|
Wang Z, Sun D, Chen YJ, Xie X, Shi Y, Tabar V, Brennan CW, Bale TA, Jayewickreme CD, Laks DR, Alcantara Llaguno S, Parada LF. Cell Lineage-Based Stratification for Glioblastoma. Cancer Cell 2020; 38:366-379.e8. [PMID: 32649888 PMCID: PMC7494533 DOI: 10.1016/j.ccell.2020.06.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/21/2020] [Accepted: 06/02/2020] [Indexed: 12/29/2022]
Abstract
Glioblastoma, the predominant adult malignant brain tumor, has been computationally classified into molecular subtypes whose functional relevance remains to be comprehensively established. Tumors from genetically engineered glioblastoma mouse models initiated by identical driver mutations in distinct cells of origin portray unique transcriptional profiles reflective of their respective lineage. Here, we identify corresponding transcriptional profiles in human glioblastoma and describe patient-derived xenografts with species-conserved subtype-discriminating functional properties. The oligodendrocyte lineage-associated glioblastoma subtype requires functional ERBB3 and harbors unique therapeutic sensitivities. These results highlight the importance of cell lineage in glioblastoma independent of driver mutations and provide a methodology for functional glioblastoma classification for future clinical investigations.
Collapse
Affiliation(s)
- Zilai Wang
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daochun Sun
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu-Jung Chen
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xuanhua Xie
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yufeng Shi
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Viviane Tabar
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cameron W Brennan
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tejus A Bale
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chenura D Jayewickreme
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Dan R Laks
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sheila Alcantara Llaguno
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Luis F Parada
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
13
|
Jung E, Alfonso J, Monyer H, Wick W, Winkler F. Neuronal signatures in cancer. Int J Cancer 2020; 147:3281-3291. [PMID: 32510582 DOI: 10.1002/ijc.33138] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Despite advances in the treatment of solid tumors, the prognosis of patients with many cancers remains poor, particularly of those with primary and metastatic brain tumors. In the last years, "Cancer Neuroscience" emerged as novel field of research at the crossroads of oncology and classical neuroscience. In primary brain tumors, including glioblastoma (GB), communicating networks that render tumor cells resistant against cytotoxic therapies were identified. To build these networks, GB cells extend neurite-like protrusions called tumor microtubes (TMs). Synapses on TMs allow tumor cells to retrieve neuronal input that fosters growth. Single cell sequencing further revealed that primary brain tumors recapitulate many steps of neurodevelopment. Interestingly, neuronal characteristics, including the ability to extend neurite-like protrusions, neuronal gene expression signatures and interactions with neurons, have now been found not only in brain and neuroendocrine tumors but also in some cancers of epithelial origin. In this review, we will provide an overview about neurite-like protrusions as well as neurodevelopmental origins, hierarchies and gene expression signatures in cancer. We will also discuss how "Cancer Neuroscience" might provide a framework for the development of novel therapies.
Collapse
Affiliation(s)
- Erik Jung
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julieta Alfonso
- Department of Clinical Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Clinical Neurobiology, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
14
|
Felker J, Broniscer A. Improving long-term survival in diffuse intrinsic pontine glioma. Expert Rev Neurother 2020; 20:647-658. [PMID: 32543245 DOI: 10.1080/14737175.2020.1775584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) is an almost universally fatal pediatric brain cancer. There has been no improvement in event-free survival (EFS) or overall survival (OS) despite immense effort through a multitude of clinical trials to find a cure. Recently, there has been a surge in the knowledge of DIPG biology, including the discovery of a recurrent H3F3A mutation in over 80% of these tumors. AREAS COVERED The authors review the most recent approaches to diagnosis and treatment of DIPG including chemotherapy, biologics, surgical approaches, and immunotherapy. EXPERT OPINION The authors propose four main opportunities to improve long-term survival. First, patients should be enrolled in scientifically sound clinical trials that include molecularly profiling either via stereotactic biopsy or liquid biopsy. Second, clinical trials should include more innovative endpoints other than traditional EFS and OS such as MRI/PET imaging findings combined with surrogates of activity (e.g. serial liquid biopsies) to better ascertain biologically active treatments. Third, innovative clinical trial approaches are needed to help allow for the rapid development of combination therapies to be tested. Finally, effort should be concentrated on reversing the effects of the histone mutation, as this malfunctioning development program seems to be key to DIPG relentlessness.
Collapse
Affiliation(s)
- James Felker
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| | - Alberto Broniscer
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| |
Collapse
|
15
|
Fortin J, Tian R, Zarrabi I, Hill G, Williams E, Sanchez-Duffhues G, Thorikay M, Ramachandran P, Siddaway R, Wong JF, Wu A, Apuzzo LN, Haight J, You-Ten A, Snow BE, Wakeham A, Goldhamer DJ, Schramek D, Bullock AN, Dijke PT, Hawkins C, Mak TW. Mutant ACVR1 Arrests Glial Cell Differentiation to Drive Tumorigenesis in Pediatric Gliomas. Cancer Cell 2020; 37:308-323.e12. [PMID: 32142668 PMCID: PMC7105820 DOI: 10.1016/j.ccell.2020.02.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/02/2019] [Accepted: 02/04/2020] [Indexed: 12/30/2022]
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors for which there is currently no effective treatment. Some of these tumors combine gain-of-function mutations in ACVR1, PIK3CA, and histone H3-encoding genes. The oncogenic mechanisms of action of ACVR1 mutations are currently unknown. Using mouse models, we demonstrate that Acvr1G328V arrests the differentiation of oligodendroglial lineage cells, and cooperates with Hist1h3bK27M and Pik3caH1047R to generate high-grade diffuse gliomas. Mechanistically, Acvr1G328V upregulates transcription factors which control differentiation and DIPG cell fitness. Furthermore, we characterize E6201 as a dual inhibitor of ACVR1 and MEK1/2, and demonstrate its efficacy toward tumor cells in vivo. Collectively, our results describe an oncogenic mechanism of action for ACVR1 mutations, and suggest therapeutic strategies for DIPGs.
Collapse
MESH Headings
- Activin Receptors, Type I/antagonists & inhibitors
- Activin Receptors, Type I/chemistry
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Animals
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Brain Neoplasms/drug therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/pathology
- Cell Differentiation/genetics
- Cell Line, Tumor
- Class I Phosphatidylinositol 3-Kinases/genetics
- Class I Phosphatidylinositol 3-Kinases/metabolism
- Female
- Glioma/drug therapy
- Glioma/genetics
- Glioma/pathology
- Histones/genetics
- Histones/metabolism
- Humans
- Lactones/pharmacology
- Male
- Mice, Transgenic
- Mutation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neuroglia/metabolism
- Neuroglia/pathology
- Oligodendroglia/pathology
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- SOXC Transcription Factors/genetics
- SOXC Transcription Factors/metabolism
Collapse
Affiliation(s)
- Jerome Fortin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Ruxiao Tian
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ida Zarrabi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Graham Hill
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Eleanor Williams
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, P.O. Box 9600 RC, Leiden, the Netherlands
| | - Midory Thorikay
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, P.O. Box 9600 RC, Leiden, the Netherlands
| | | | - Robert Siddaway
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G1X8, Canada
| | - Jong Fu Wong
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Annette Wu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Lorraine N Apuzzo
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06268, USA
| | - Jillian Haight
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Annick You-Ten
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Bryan E Snow
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Andrew Wakeham
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06268, USA
| | - Daniel Schramek
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alex N Bullock
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, P.O. Box 9600 RC, Leiden, the Netherlands
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G1X8, Canada; Division of Pathology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
16
|
Mendez FM, Núñez FJ, Garcia-Fabiani MB, Haase S, Carney S, Gauss JC, Becher OJ, Lowenstein PR, Castro MG. Epigenetic reprogramming and chromatin accessibility in pediatric diffuse intrinsic pontine gliomas: a neural developmental disease. Neuro Oncol 2020; 22:195-206. [PMID: 32078691 PMCID: PMC7032633 DOI: 10.1093/neuonc/noz218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but deadly pediatric brainstem tumor. To date, there is no effective therapy for DIPG. Transcriptomic analyses have revealed DIPGs have a distinct profile from other pediatric high-grade gliomas occurring in the cerebral hemispheres. These unique genomic characteristics coupled with the younger median age group suggest that DIPG has a developmental origin. The most frequent mutation in DIPG is a lysine to methionine (K27M) mutation that occurs on H3F3A and HIST1H3B/C, genes encoding histone variants. The K27M mutation disrupts methylation by polycomb repressive complex 2 on histone H3 at lysine 27, leading to global hypomethylation. Histone 3 lysine 27 trimethylation is an important developmental regulator controlling gene expression. This review discusses the developmental and epigenetic mechanisms driving disease progression in DIPG, as well as the profound therapeutic implications of epigenetic programming.
Collapse
Affiliation(s)
- Flor M Mendez
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Felipe J Núñez
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria B Garcia-Fabiani
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Santiago Haase
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Stephen Carney
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jessica C Gauss
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Oren J Becher
- Department of Pediatrics, Northwestern University, Chicago, Illinois
- Ann & Robert Lurie Children’s Hospital of Chicago, Division of Hematology-Oncology and Stem Cell Transplant, Chicago, Illinois
| | - Pedro R Lowenstein
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria G Castro
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
17
|
RBM3 promotes neurogenesis in a niche-dependent manner via IMP2-IGF2 signaling pathway after hypoxic-ischemic brain injury. Nat Commun 2019; 10:3983. [PMID: 31484925 PMCID: PMC6726629 DOI: 10.1038/s41467-019-11870-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Hypoxic ischemia (HI) is an acute brain threat across all age groups. Therapeutic hypothermia ameliorates resulting injury in neonates but its side effects prevent routine use in adults. Hypothermia up-regulates a small protein subset that includes RNA-binding motif protein 3 (RBM3), which is neuroprotective under stressful conditions. Here we show how RBM3 stimulates neuronal differentiation and inhibits HI-induced apoptosis in the two areas of persistent adult neurogenesis, the subventricular zone (SVZ) and the subgranular zone (SGZ), while promoting neural stem/progenitor cell (NSPC) proliferation after HI injury only in the SGZ. RBM3 interacts with IGF2 mRNA binding protein 2 (IMP2), elevates its expression and thereby stimulates IGF2 release in SGZ but not SVZ-NSPCs. In summary, we describe niche-dependent regulation of neurogenesis after adult HI injury via the novel RBM3-IMP2-IGF2 signaling pathway. Therapeutic hypothermia is a potent tool in the treatment of neonatal hypoxic-ischemic (HI) injury, yet the underlying mechanisms remain unclear. Here, authors demonstrate how the RNA-binding motif protein RBM3, which is induced by mild cooling while global translation rate is slowed down, contributes substantially to neuroregeneration after adult HI injury, specifically in the subventricular zone and subgranular zone.
Collapse
|
18
|
Sorrells SF, Paredes MF, Velmeshev D, Herranz-Pérez V, Sandoval K, Mayer S, Chang EF, Insausti R, Kriegstein AR, Rubenstein JL, Manuel Garcia-Verdugo J, Huang EJ, Alvarez-Buylla A. Immature excitatory neurons develop during adolescence in the human amygdala. Nat Commun 2019; 10:2748. [PMID: 31227709 PMCID: PMC6588589 DOI: 10.1038/s41467-019-10765-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
The human amygdala grows during childhood, and its abnormal development is linked to mood disorders. The primate amygdala contains a large population of immature neurons in the paralaminar nuclei (PL), suggesting protracted development and possibly neurogenesis. Here we studied human PL development from embryonic stages to adulthood. The PL develops next to the caudal ganglionic eminence, which generates inhibitory interneurons, yet most PL neurons express excitatory markers. In children, most PL cells are immature (DCX+PSA-NCAM+), and during adolescence many transition into mature (TBR1+VGLUT2+) neurons. Immature PL neurons persist into old age, yet local progenitor proliferation sharply decreases in infants. Using single nuclei RNA sequencing, we identify the transcriptional profile of immature excitatory neurons in the human amygdala between 4-15 years. We conclude that the human PL contains excitatory neurons that remain immature for decades, a possible substrate for persistent plasticity at the interface of the hippocampus and amygdala.
Collapse
Affiliation(s)
- Shawn F Sorrells
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Dmitry Velmeshev
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
- Predepartamental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I, 12071, Castelló de la Plana, Spain
| | - Kadellyn Sandoval
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Simone Mayer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Ricardo Insausti
- Human Neuroanatomy Laboratory, School of Medicine and CRIB, University of Castilla-La Mancha, 02006, Albacete, Spain
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - John L Rubenstein
- Department of Psychiatry, Rock Hall, University of California, San Francisco, San Francisco, CA, 94158-2324, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
19
|
H3.3 K27M depletion increases differentiation and extends latency of diffuse intrinsic pontine glioma growth in vivo. Acta Neuropathol 2019; 137:637-655. [PMID: 30770999 DOI: 10.1007/s00401-019-01975-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/22/2019] [Accepted: 02/08/2019] [Indexed: 12/30/2022]
Abstract
Histone H3 K27M mutation is the defining molecular feature of the devastating pediatric brain tumor, diffuse intrinsic pontine glioma (DIPG). The prevalence of histone H3 K27M mutations indicates a critical role in DIPGs, but the contribution of the mutation to disease pathogenesis remains unclear. We show that knockdown of this mutation in DIPG xenografts restores K27M-dependent loss of H3K27me3 and delays tumor growth. Comparisons of matched DIPG xenografts with and without K27M knockdown allowed identification of mutation-specific effects on the transcriptome and epigenome. The resulting transcriptional changes recapitulate expression signatures from K27M primary DIPG tumors and are strongly enriched for genes associated with nervous system development. Integrated analysis of ChIP-seq and expression data showed that genes upregulated by the mutation are overrepresented in apparently bivalent promoters. Many of these targets are associated with more immature differentiation states. Expression profiles indicate K27M knockdown decreases proliferation and increases differentiation within lineages represented in DIPG. These data suggest that K27M-mediated loss of H3K27me3 directly regulates a subset of genes by releasing poised promoters, and contributes to tumor phenotype and growth by limiting differentiation. The delayed tumor growth associated with knockdown of H3 K27M provides evidence that this highly recurrent mutation is a relevant therapeutic target.
Collapse
|
20
|
Larson JD, Kasper LH, Paugh BS, Jin H, Wu G, Kwon CH, Fan Y, Shaw TI, Silveira AB, Qu C, Xu R, Zhu X, Zhang J, Russell HR, Peters JL, Finkelstein D, Xu B, Lin T, Tinkle CL, Patay Z, Onar-Thomas A, Pounds SB, McKinnon PJ, Ellison DW, Zhang J, Baker SJ. Histone H3.3 K27M Accelerates Spontaneous Brainstem Glioma and Drives Restricted Changes in Bivalent Gene Expression. Cancer Cell 2019; 35:140-155.e7. [PMID: 30595505 PMCID: PMC6570409 DOI: 10.1016/j.ccell.2018.11.015] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/13/2018] [Accepted: 11/23/2018] [Indexed: 12/31/2022]
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are incurable childhood brainstem tumors with frequent histone H3 K27M mutations and recurrent alterations in PDGFRA and TP53. We generated genetically engineered inducible mice and showed that H3.3 K27M enhanced neural stem cell self-renewal while preserving regional identity. Neonatal induction of H3.3 K27M cooperated with activating platelet-derived growth factor receptor α (PDGFRα) mutant and Trp53 loss to accelerate development of diffuse brainstem gliomas that recapitulated human DIPG gene expression signatures and showed global changes in H3K27 posttranslational modifications, but relatively restricted gene expression changes. Genes upregulated in H3.3 K27M tumors were enriched for those associated with neural development where H3K27me3 loss released the poised state of apparently bivalent promoters, whereas downregulated genes were enriched for those encoding homeodomain transcription factors.
Collapse
Affiliation(s)
- Jon D Larson
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lawryn H Kasper
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Barbara S Paugh
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hongjian Jin
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Gang Wu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chang-Hyuk Kwon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Timothy I Shaw
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - André B Silveira
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chunxu Qu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Raymond Xu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiaoyan Zhu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Junyuan Zhang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Helen R Russell
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer L Peters
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tong Lin
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zoltan Patay
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanley B Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter J McKinnon
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David W Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
21
|
Grill J, Debily MA, Varlet P, Plessier A, Le Dret L, Beccaria K, Puget S, Castel D. The dark matter of diffuse intrinsic pontine gliomas: an update. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2019.1560262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Jacques Grill
- Equipe Nouvelles Thérapeutiques Anticancéreuses, Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique, Gustave Roussy, Université Paris-Saclay, Villejuif cedex, France
- Departement de Cancerologie de l’Enfant et de l’Adolescent, Gustave Roussy, Villejuif cedex, France
| | - Marie-Anne Debily
- Equipe Nouvelles Thérapeutiques Anticancéreuses, Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique, Gustave Roussy, Université Paris-Saclay, Villejuif cedex, France
- Department Master Biologie et Sante, Universite Evry Val d'Essonne, Evry cedex, France
| | - Pascale Varlet
- Service de Neuropathologie, Hôpital Sainte-Anne, Paris cedex, France
| | - Alexandre Plessier
- Equipe Nouvelles Thérapeutiques Anticancéreuses, Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique, Gustave Roussy, Université Paris-Saclay, Villejuif cedex, France
| | - Ludivine Le Dret
- Equipe Nouvelles Thérapeutiques Anticancéreuses, Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique, Gustave Roussy, Université Paris-Saclay, Villejuif cedex, France
| | - Kevin Beccaria
- Departement de Cancerologie de l’Enfant et de l’Adolescent, Gustave Roussy, Villejuif cedex, France
- Service de Neurochirurgie Pédiatrique, Hôpital Necker - Enfants Malades, Paris cedex, France
| | - Stéphanie Puget
- Service de Neurochirurgie Pédiatrique, Hôpital Necker - Enfants Malades, Paris cedex, France
| | - David Castel
- Equipe Nouvelles Thérapeutiques Anticancéreuses, Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique, Gustave Roussy, Université Paris-Saclay, Villejuif cedex, France
| |
Collapse
|
22
|
Intersection of Brain Development and Paediatric Diffuse Midline Gliomas: Potential Role of Microenvironment in Tumour Growth. Brain Sci 2018; 8:brainsci8110200. [PMID: 30453529 PMCID: PMC6266894 DOI: 10.3390/brainsci8110200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/03/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a devastating and incurable paediatric brain tumour with a median overall survival of 9 months. Until recently, DIPGs were treated similarly to adult gliomas, but due to the advancement in molecular and imaging technologies, our understanding of these tumours has increased dramatically. While extensive research is being undertaken to determine the function of the molecular aberrations in DIPG, there are significant gaps in understanding the biology and the influence of the tumour microenvironment on DIPG growth, specifically in regards to the developing pons. The precise orchestration and co-ordination of the development of the brain, the most complex organ in the body, is still not fully understood. Herein, we present a brief overview of brainstem development, discuss the developing microenvironment in terms of DIPG growth, and provide a basis for the need for studies focused on bridging pontine development and DIPG microenvironment. Conducting investigations in the context of a developing brain will lead to a better understanding of the role of the tumour microenvironment and will help lead to identification of drivers of tumour growth and therapeutic resistance.
Collapse
|
23
|
Hoeman C, Shen C, Becher OJ. CDK4/6 and PDGFRA Signaling as Therapeutic Targets in Diffuse Intrinsic Pontine Glioma. Front Oncol 2018; 8:191. [PMID: 29904623 PMCID: PMC5990603 DOI: 10.3389/fonc.2018.00191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are incurable childhood brain tumors, whereby the standard of care is focal radiation, a treatment that provides temporary relief for most patients. Surprisingly, decades of clinical trials have failed to identify additional therapies that can prolong survival in this disease. In this conference manuscript, we discuss how genetic engineered mouse modeling techniques with the use of a retroviral gene delivery system can help dissect the complex pathophysiology of this disease. With this approach, autochthonous murine DIPG models can be readily induced to (1) help interrogate the function of novel genetic alterations in tumorigenesis, (2) identify candidate cells of origin for this disease, (3) address how region-specific differences in the central nervous system influence the process of gliomagenesis, and (4) evaluate novel therapeutics in an immunocompetent model.
Collapse
Affiliation(s)
- Christine Hoeman
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
| | - Chen Shen
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
| | - Oren J Becher
- Department of Pediatrics, Northwestern University, Chicago, IL, United States.,Ann & Robert Lurie Children's Hospital of Chicago, Division of Hematology-Oncology and Stem Cell Transplant, Chicago, IL, United States
| |
Collapse
|
24
|
Han HJ, Jain P, Resnick AC. Shared ACVR1 mutations in FOP and DIPG: Opportunities and challenges in extending biological and clinical implications across rare diseases. Bone 2018; 109:91-100. [PMID: 28780023 PMCID: PMC7888549 DOI: 10.1016/j.bone.2017.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Gain-of-function mutations in the Type I Bone Morphogenic Protein (BMP) receptor ACVR1 have been identified in two diseases: Fibrodysplasia Ossificans Progressiva (FOP), a rare autosomal dominant disorder characterized by genetically driven heterotopic ossification, and in 20-25% of Diffuse Intrinsic Pontine Gliomas (DIPGs), a pediatric brain tumor with no effective therapies and dismal median survival. While the ACVR1 mutation is causal for FOP, its role in DIPG tumor biology remains under active investigation. Here, we discuss cross-fertilization between the FOP and DIPG fields, focusing on the biological mechanisms and principles gleaned from FOP that can be applied to DIPG biology. We highlight our current knowledge of ACVR1 in both diseases, and then describe the growing opportunities and barriers to effectively investigate ACVR1 in DIPG. Importantly, learning from other seemingly unrelated diseases harboring similar mutations may uncover novel mechanisms or processes for future investigation.
Collapse
Affiliation(s)
- Harry J Han
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States
| | - Payal Jain
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States
| | - Adam C Resnick
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States.
| |
Collapse
|
25
|
Bouchat J, Couturier B, Marneffe C, Gankam-Kengne F, Balau B, De Swert K, Brion JP, Poncelet L, Gilloteaux J, Nicaise C. Regional oligodendrocytopathy and astrocytopathy precede myelin loss and blood-brain barrier disruption in a murine model of osmotic demyelination syndrome. Glia 2017; 66:606-622. [DOI: 10.1002/glia.23268] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/28/2022]
Affiliation(s)
| | - Bruno Couturier
- Department of General Medicine; Erasme Hospital, Université Libre de Bruxelles; Bruxelles Belgium
- Laboratory of Histology, Neuroanatomy and Neuropathology; Université Libre de Bruxelles; Bruxelles Belgium
| | | | - Fabrice Gankam-Kengne
- Laboratory of Histology, Neuroanatomy and Neuropathology; Université Libre de Bruxelles; Bruxelles Belgium
- Department of Nephrology; EpiCURA Ath; Ath Belgium
| | - Benoît Balau
- URPhyM - NARILIS, Université de Namur; Namur Belgium
| | | | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology; Université Libre de Bruxelles; Bruxelles Belgium
| | - Luc Poncelet
- Laboratory of Anatomy, Biomechanics and Organogenesis; Université Libre de Bruxelles; Bruxelles Belgium
| | - Jacques Gilloteaux
- URPhyM - NARILIS, Université de Namur; Namur Belgium
- Department of Anatomical Sciences; St George's University School of Medicine, Newcastle upon Tyne; United Kingdom
| | | |
Collapse
|
26
|
Kratochwil CF, Maheshwari U, Rijli FM. The Long Journey of Pontine Nuclei Neurons: From Rhombic Lip to Cortico-Ponto-Cerebellar Circuitry. Front Neural Circuits 2017; 11:33. [PMID: 28567005 PMCID: PMC5434118 DOI: 10.3389/fncir.2017.00033] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/28/2017] [Indexed: 01/26/2023] Open
Abstract
The pontine nuclei (PN) are the largest of the precerebellar nuclei, neuronal assemblies in the hindbrain providing principal input to the cerebellum. The PN are predominantly innervated by the cerebral cortex and project as mossy fibers to the cerebellar hemispheres. Here, we comprehensively review the development of the PN from specification to migration, nucleogenesis and circuit formation. PN neurons originate at the posterior rhombic lip and migrate tangentially crossing several rhombomere derived territories to reach their final position in ventral part of the pons. The developing PN provide a classical example of tangential neuronal migration and a study system for understanding its molecular underpinnings. We anticipate that understanding the mechanisms of PN migration and assembly will also permit a deeper understanding of the molecular and cellular basis of cortico-cerebellar circuit formation and function.
Collapse
Affiliation(s)
- Claudius F Kratochwil
- Chair in Zoology and Evolutionary Biology, Department of Biology, University of KonstanzKonstanz, Germany.,Zukunftskolleg, University of KonstanzKonstanz, Germany
| | - Upasana Maheshwari
- Friedrich Miescher Institute for Biomedical ResearchBasel, Switzerland.,University of BaselBasel, Switzerland
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical ResearchBasel, Switzerland.,University of BaselBasel, Switzerland
| |
Collapse
|