1
|
Zhang Q, Huang J, Chen X, Li L, Chen L, Zhou X, Zhao X, Liu M, Zhao W, Yan J, Wang Y, Su Y, Liu Y, Xu S, Zeng W. Mesenchymal Stem Cell-Derived Mitochondrial Transfer Promotes Tip Cell Phenotype via Glutathione Metabolic Reprogramming in Stroke Mice. ACS NANO 2025. [PMID: 40448640 DOI: 10.1021/acsnano.4c15759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Angiogenesis is crucial to improving neurovascular remodeling poststroke. Therein, the transformation of endothelial cells (ECs) to tip cells is essential in initiating angiogenesis. Mitochondrial damage in ECs poststroke and associated metabolic disorder are key factors repressing angiogenesis, but the mechanisms are unknown. Here, we designed an Arg-Gly-Asp peptide (RGD)-modified, mitochondria-enriched, and extracellular vesicle mimetics (mitoEVMs) platform for mitochondrial transfer. RGD mediated the mesenchymal stem cell-derived mitochondria transfer to ECs around the lesion targetedly. We found MSC-derived mitochondria promoted tip cell transition and further stimulated angiogenesis after stroke, alleviated brain atrophy, and improved functional rehabilitation. We noticed mitochondrial transfer rescued mitochondrial function in ECs and reprogrammed glutathione metabolism to activate the mTORC1 pathway, upregulated the expression of p4E-BP1 and VEGFR2, and ultimately facilitated tip cell transition. Our work elucidates the mechanism of MSC-derived mitochondrial transfer in poststroke treatment and proposes a potential approach for rehabilitation after stroke.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
- Department of Pain and Rehabilitation, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiaxin Huang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xi Chen
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lang Li
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Lin Chen
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xin Zhou
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xingli Zhao
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Min Liu
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Wenyan Zhao
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Juan Yan
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yueying Wang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yang Su
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yong Liu
- Department of Pain and Rehabilitation, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shangcheng Xu
- School of public health, Chongqing Medical University, Chongqing 400016, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400038, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
2
|
He T, Zhang M, Qin J, Wang Y, Li S, Du C, Jiao J, Ji F. Endothelial PD-1 Regulates Vascular Homeostasis and Oligodendrogenesis during Brain Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417410. [PMID: 40013943 PMCID: PMC12021089 DOI: 10.1002/advs.202417410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Appropriate vascular and neural development is essential for central nervous system (CNS). Although programmed cell death receptor 1 (PD-1) mediates neurogenesis, its role in cerebrovascular development remains poorly understood. Here, a correlation between cerebral vessels and oligodendrocyte precursor cells (OPCs) is revealed during brain development. The ablation of endothelial PD-1 triggers cortical hypervascularization through excessive angiogenic sprouting, concomitantly driving OPC differentiation. These alterations disrupt blood brain barrier (BBB) maturation, induce dysmyelination, and ultimately result in abnormal behavior in mice. Mechanistically, the loss of endothelial PD-1 suppresses the activity of the Wnt/β-catenin signaling pathway, thereby disrupting normal angiogenesis. Concurrently, it activates the MEK1/2-ERK1/2-GLI1 pathway, leading to increased GREMLIN1 (GREM1) expression. Elevated GREM1 secretion inhibits the BMP/SMAD1/5/SMAD4 signaling cascade in OPCs, which inhibits oligodendrogenesis and myelination. These findings indicate the importance of endothelial cell-intrinsic PD-1 in regulating the oligovascular niche, and suggest potential therapeutic implications for neurological disorders associated with disrupted vascular development.
Collapse
Affiliation(s)
- Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100190China
| | - Mengtian Zhang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanyan Wang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Sihan Li
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chaoyi Du
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
3
|
Ruhoff VT, Leijnse N, Doostmohammadi A, Bendix PM. Filopodia: integrating cellular functions with theoretical models. Trends Cell Biol 2025; 35:129-140. [PMID: 38969554 DOI: 10.1016/j.tcb.2024.05.005] [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: 02/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 07/07/2024]
Abstract
Filopodia, widely distributed on cell surfaces, are distinguished by their dynamic extensions, playing pivotal roles in a myriad of biological processes. Their functions span from mechanosensing and guidance to cell-cell communication during cellular organization in the early embryo. Filopodia have significant roles in pathogenic processes, such as cancer invasion and viral dissemination. Molecular mapping of the filopodome has revealed generic components essential for filopodia functions. In parallel, recent insights into biophysical mechanisms governing filopodia dynamics have provided the foundation for broader investigations of filopodia's biological functions. We highlight recent discoveries of engagement of filopodia in various stages of development and pathogenesis and present an overview of intricate molecular and physical features of these cellular structures across a spectrum of cellular activities.
Collapse
Affiliation(s)
| | - Natascha Leijnse
- Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| | - Amin Doostmohammadi
- Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| | - Poul Martin Bendix
- Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark.
| |
Collapse
|
4
|
Luo Y, Zhang Y, Feng Y, Zeng X, Zhu D, Yang Y, Hu H, Wang Q, Guo L, Zou L, Zhong X. Prenatal exposure to low doses of benzophenone-3 elicits disruption of cortical vasculature in fetuses through perturbations in Wnt/β-catenin signaling correlating with depression-like behavior in offspring mice. Toxicology 2024; 509:153960. [PMID: 39343157 DOI: 10.1016/j.tox.2024.153960] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Benzophenone-3 (BP-3), commonly used in personal care products, is routinely detected in environmental and human matrices. Evidence delineates a correlation between gestational BP-3 exposure and emotional and social disorders in children and adolescents. However, sensitive target cells and the mode of action underlying the early responses to environmentally relevant level of BP-3 exposure remain unclear. In this study, 0.3 and 3 mg/kg of BP-3 were administered to pregnant mice. Compared with the control group, the cortical blood vessel development process manifested the highest susceptibility to BP-3 exposure using transcriptomic sequencing at embryonic day 14 (E14). Notably, the diminution in vascular density and tight junction proteins presence was observed in the fetal cortex at E14, concomitant with the suppressed transcriptional activity of genes essential to angiogenesis and barrier formation. Strikingly, the investigation revealed that BP-3 exposure impeded vascular sprouting in aortic ring explants and neuroendothelial migration, implicating the Wnt/β-catenin signaling pathway. Moreover, BP-3 exposure compromised perivascular neural stem cell differentiation. Cortical vascular injury correlated with the exhibition of depression-like behavior in four-week postnatal progeny. These insights underscore the cerebrovasculature as an early sensitive target for low doses of BP-3 exposure, fostering the development of biomarkers and the establishment of the adverse outcome pathway framework for BP-3 hazard evaluation.
Collapse
Affiliation(s)
- Yijun Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yangjian Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yang Feng
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China
| | - Xiangyu Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Dan Zhu
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China
| | - Ying Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Haichen Hu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lianhong Zou
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China; Geriatric Immunization Research Center of Hunan Provincial Geriatric Institute, Changsha, Hunan, China.
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
5
|
Chen J, Crouch EE, Zawadzki ME, Jacobs KA, Mayo LN, Choi JJY, Lin PY, Shaikh S, Tsui J, Gonzalez-Granero S, Waller S, Kelekar A, Kang G, Valenzuela EJ, Birrueta JO, Diafos LN, Wedderburn-Pugh K, Di Marco B, Xia W, Han CZ, Coufal NG, Glass CK, Fancy SPJ, Alfonso J, Kriegstein AR, Oldham MC, Garcia-Verdugo JM, Kutys ML, Lehtinen MK, Combes AJ, Huang EJ. Proinflammatory immune cells disrupt angiogenesis and promote germinal matrix hemorrhage in prenatal human brain. Nat Neurosci 2024; 27:2115-2129. [PMID: 39349662 PMCID: PMC11537974 DOI: 10.1038/s41593-024-01769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 08/19/2024] [Indexed: 10/28/2024]
Abstract
Germinal matrix hemorrhage (GMH) is a devastating neurodevelopmental condition affecting preterm infants, but why blood vessels in this brain region are vulnerable to rupture remains unknown. Here we show that microglia in prenatal mouse and human brain interact with nascent vasculature in an age-dependent manner and that ablation of these cells in mice reduces angiogenesis in the ganglionic eminences, which correspond to the human germinal matrix. Consistent with these findings, single-cell transcriptomics and flow cytometry show that distinct subsets of CD45+ cells from control preterm infants employ diverse signaling mechanisms to promote vascular network formation. In contrast, CD45+ cells from infants with GMH harbor activated neutrophils and monocytes that produce proinflammatory factors, including azurocidin 1, elastase and CXCL16, to disrupt vascular integrity and cause hemorrhage in ganglionic eminences. These results underscore the brain's innate immune cells in region-specific angiogenesis and how aberrant activation of these immune cells promotes GMH in preterm infants.
Collapse
Affiliation(s)
- Jiapei Chen
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Elizabeth E Crouch
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Miriam E Zawadzki
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
- Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Kyle A Jacobs
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Lakyn N Mayo
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- UCSF-UC Berkeley Joint Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Ja-Yoon Choi
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Pin-Yeh Lin
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Saba Shaikh
- UCSF CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Jessica Tsui
- UCSF CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Susana Gonzalez-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, Valencia, Spain
| | - Shamari Waller
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Avani Kelekar
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Gugene Kang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Edward J Valenzuela
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Janeth Ochoa Birrueta
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Loukas N Diafos
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Kaylee Wedderburn-Pugh
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | - Barbara Di Marco
- Department of Clinical Neurobiology, University Hospital Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Wenlong Xia
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nicole G Coufal
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Stephen P J Fancy
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Julieta Alfonso
- Department of Clinical Neurobiology, University Hospital Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Arnold R Kriegstein
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael C Oldham
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, Valencia, Spain
| | - Matthew L Kutys
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- UCSF-UC Berkeley Joint Graduate Program in Bioengineering, University of California San Francisco, San Francisco, CA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Alexis J Combes
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- Department of Pathology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA.
- Pathology Service 113B, San Francisco VA Health Care Systems, San Francisco, CA, USA.
| |
Collapse
|
6
|
Augustin HG, Koh GY. A systems view of the vascular endothelium in health and disease. Cell 2024; 187:4833-4858. [PMID: 39241746 DOI: 10.1016/j.cell.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 09/09/2024]
Abstract
The dysfunction of blood-vessel-lining endothelial cells is a major cause of mortality. Although endothelial cells, being present in all organs as a single-cell layer, are often conceived as a rather inert cell population, the vascular endothelium as a whole should be considered a highly dynamic and interactive systemically disseminated organ. We present here a holistic view of the field of vascular research and review the diverse functions of blood-vessel-lining endothelial cells during the life cycle of the vasculature, namely responsive and relaying functions of the vascular endothelium and the responsive roles as instructive gatekeepers of organ function. Emerging translational perspectives in regenerative medicine, preventive medicine, and aging research are developed. Collectively, this review is aimed at promoting disciplinary coherence in the field of angioscience for a broader appreciation of the importance of the vasculature for organ function, systemic health, and healthy aging.
Collapse
Affiliation(s)
- Hellmut G Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
7
|
Murayama F, Asai H, Patra AK, Wake H, Miyata T, Hattori Y. A novel preparation for histological analyses of intraventricular macrophages in the embryonic brain. Dev Growth Differ 2024; 66:329-337. [PMID: 38894655 PMCID: PMC11457502 DOI: 10.1111/dgd.12935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Microglia colonize the brain starting on embryonic day (E) 9.5 in mice, and their population increases with development. We have previously demonstrated that some microglia are derived from intraventricular macrophages, which frequently infiltrate the pallium at E12.5. To address how the infiltration of intraventricular macrophages is spatiotemporally regulated, histological analyses detecting how these cells associate with the surrounding cells at the site of infiltration into the pallial surface are essential. Using two-photon microscopy-based in vivo imaging, we demonstrated that most intraventricular macrophages adhere to the ventricular surface. This is a useful tool for imaging intraventricular macrophages maintaining their original position, but this method cannot be used for observing deeper brain regions. Meanwhile, we found that conventional cryosection-based and naked pallial slice-based observation resulted in unexpected detachment from the ventricular surface of intraventricular macrophages and their mislocation, suggesting that previous histological analyses might have failed to determine their physiological number and location in the ventricular space. To address this, we sought to establish a methodological preparation that enables us to delineate the structure and cellular interactions when intraventricular macrophages infiltrate the pallium. Here, we report that brain slices pretreated with agarose-embedding maintained adequate density and proper positioning of intraventricular macrophages on the ventricular surface. This method also enabled us to perform the immunostaining. We believe that this is helpful for conducting histological analyses to elucidate the mechanisms underlying intraventricular macrophage infiltration into the pallium and their cellular properties, leading to further understanding of the process of microglial colonization into the developing brain.
Collapse
Affiliation(s)
- Futoshi Murayama
- Department of Anatomy and Cell BiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hisa Asai
- Department of Anatomy and Cell BiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Arya Kirone Patra
- Department of Anatomy and Cell BiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hiroaki Wake
- Department of Anatomy and Molecular BiologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of Physiological Sciences, Graduate School for Advanced StudiesSOKENDAIHayamaJapan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological SciencesNational Institute of Natural SciencesOkazakiJapan
| | - Takaki Miyata
- Department of Anatomy and Cell BiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yuki Hattori
- Department of Anatomy and Cell BiologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
8
|
Stepien BK, Wielockx B. From Vessels to Neurons-The Role of Hypoxia Pathway Proteins in Embryonic Neurogenesis. Cells 2024; 13:621. [PMID: 38607059 PMCID: PMC11012138 DOI: 10.3390/cells13070621] [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: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.
Collapse
Affiliation(s)
- Barbara K. Stepien
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Experimental Centre, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
9
|
Zhu H, Du Z, Lu R, Zhou Q, Shen Y, Jiang Y. Investigating the Mechanism of Chufan Yishen Formula in Treating Depression through Network Pharmacology and Experimental Verification. ACS OMEGA 2024; 9:12698-12710. [PMID: 38524447 PMCID: PMC10955564 DOI: 10.1021/acsomega.3c08350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/26/2024]
Abstract
Objective: To investigate the antidepressant effect and potential mechanism of the Chufan Yishen Formula (CFYS) through network pharmacology, molecular docking, and experimental verification. Methods: The active ingredients and their target genes of CFYS were identified through Traditional Chinese Medicine Systems Pharmacology (TCMSP) and TCM-ID. We obtained the differentially expressed genes in patients with depression from the GEO database and screened out the genes intersecting with the target genes of CFYS to construct the PPI network. The key pathways were selected through STRING and KEGG. Then, molecular docking and experimental verification were performed. Results: A total of 113 effective components and 195 target genes were obtained. After intersecting the target genes with the differentially expressed genes in patients with depression, we obtained 37 differential target genes, among which HMOX1, VEGFA, etc., were the key genes. After enriching the differential target genes by KEGG, we found that the "chemical carcinogenesis-reactive oxygen species" pathway was the key pathway for the CFYS antidepressant effect. Besides, VEGFA might be a key marker for depression. Experimental verification found that CFYS could significantly improve the behavioral indicators of rats with depression models, including improving the antioxidant enzyme activity and increasing VEGFA levels. The results are consistent with the network pharmacology analysis. Conclusions: CFYS treatment for depression is a multicomponent, multitarget, and multipathway complex process, which may mainly exert an antidepressant effect by improving the neuron antioxidant stress response and regulating VEGFA levels.
Collapse
Affiliation(s)
- Haohao Zhu
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| | - Zhiqiang Du
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| | - Rongrong Lu
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| | - Qin Zhou
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| | - Yuan Shen
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| | - Ying Jiang
- Mental Health
Center of
Jiangnan University, Wuxi, Jiangsu 214151, China
| |
Collapse
|
10
|
Wilsch-Bräuninger M, Peters J, Huttner WB. High-resolution 3D ultrastructural analysis of developing mouse neocortex reveals long slender processes of endothelial cells that enter neural cells. Front Cell Dev Biol 2024; 12:1344734. [PMID: 38500687 PMCID: PMC10945550 DOI: 10.3389/fcell.2024.1344734] [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: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 03/20/2024] Open
Abstract
The development of the neocortex involves an interplay between neural cells and the vasculature. However, little is known about this interplay at the ultrastructural level. To gain a 3D insight into the ultrastructure of the developing neocortex, we have analyzed the embryonic mouse neocortex by serial block-face scanning electron microscopy (SBF-SEM). In this study, we report a first set of findings that focus on the interaction of blood vessels, notably endothelial tip cells (ETCs), and the neural cells in this tissue. A key observation was that the processes of ETCs, located either in the ventricular zone (VZ) or subventricular zone (SVZ)/intermediate zone (IZ), can enter, traverse the cytoplasm, and even exit via deep plasma membrane invaginations of the host cells, including apical progenitors (APs), basal progenitors (BPs), and newborn neurons. More than half of the ETC processes were found to enter the neural cells. Striking examples of this ETC process "invasion" were (i) protrusions of apical progenitors or newborn basal progenitors into the ventricular lumen that contained an ETC process inside and (ii) ETC process-containing protrusions of neurons that penetrated other neurons. Our observations reveal a - so far unknown - complexity of the ETC-neural cell interaction.
Collapse
Affiliation(s)
| | | | - Wieland B. Huttner
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
11
|
Chen X, Sun X, Ge Y, Zhou X, Chen JF. Targeting adenosine A 2A receptors for early intervention of retinopathy of prematurity. Purinergic Signal 2024:10.1007/s11302-024-09986-x. [PMID: 38329708 DOI: 10.1007/s11302-024-09986-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Retinopathy of prematurity (ROP) continues to pose a significant threat to the vision of numerous children worldwide, primarily owing to the increased survival rates of premature infants. The pathologies of ROP are mainly linked to impaired vascularization as a result of hyperoxia, leading to subsequent neovascularization. Existing treatments, including anti-vascular endothelial growth factor (VEGF) therapies, have thus far been limited to addressing pathological angiogenesis at advanced ROP stages, inevitably leading to adverse side effects. Intervention to promote physiological angiogenesis during the initial stages could hold the potential to prevent ROP. Adenosine A2A receptors (A2AR) have been identified in various ocular cell types, exhibiting distinct densities and functionally intricate connections with oxygen metabolism. In this review, we discuss experimental evidence that strongly underscores the pivotal role of A2AR in ROP. In particular, A2AR blockade may represent an effective treatment strategy, mitigating retinal vascular loss by reversing hyperoxia-mediated cellular proliferation inhibition and curtailing hypoxia-mediated neovascularization in oxygen-induced retinopathy (OIR). These effects stem from the interplay of endothelium, neuronal and glial cells, and novel molecular pathways (notably promoting TGF-β signaling) at the hyperoxia phase. We propose that pharmacological targeting of A2AR signaling may confer an early intervention for ROP with distinct therapeutic benefits and mechanisms than the anti-VEGF therapy.
Collapse
Affiliation(s)
- Xuhao Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaoting Sun
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Ge
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xuzhao Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China.
| |
Collapse
|
12
|
Andrews MG, Pearson CA. Toward an understanding of glucose metabolism in radial glial biology and brain development. Life Sci Alliance 2024; 7:e202302193. [PMID: 37798120 PMCID: PMC10556723 DOI: 10.26508/lsa.202302193] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
Decades of research have sought to determine the intrinsic and extrinsic mechanisms underpinning the regulation of neural progenitor maintenance and differentiation. A series of precise temporal transitions within progenitor cell populations generates all the appropriate neural cell types while maintaining a pool of self-renewing progenitors throughout embryogenesis. Recent technological advances have enabled us to gain new insights at the single-cell level, revealing an interplay between metabolic state and developmental progression that impacts the timing of proliferation and neurogenesis. This can have long-term consequences for the developing brain's neuronal specification, maturation state, and organization. Furthermore, these studies have highlighted the need to reassess the instructive role of glucose metabolism in determining progenitor cell division, differentiation, and fate. This review focuses on glucose metabolism (glycolysis) in cortical progenitor cells and the emerging focus on glycolysis during neurogenic transitions. Furthermore, we discuss how the field can learn from other biological systems to improve our understanding of the spatial and temporal changes in glycolysis in progenitors and evaluate functional neurological outcomes.
Collapse
Affiliation(s)
- Madeline G Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Caroline A Pearson
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
13
|
Eşiyok N, Heide M. The SVZ stem cell niche-components, functions, and in vitro modelling. Front Cell Dev Biol 2023; 11:1332901. [PMID: 38188021 PMCID: PMC10766702 DOI: 10.3389/fcell.2023.1332901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024] Open
Abstract
Neocortical development depends on the intrinsic ability of neural stem and progenitor cells to proliferate and differentiate to generate the different kinds of neurons in the adult brain. These progenitor cells can be distinguished into apical progenitors, which occupy a stem cell niche in the ventricular zone and basal progenitors, which occupy a stem cell niche in the subventricular zone (SVZ). During development, the stem cell niche provided in the subventricular zone enables the increased proliferation and self-renewal of basal progenitors, which likely underlie the expansion of the human neocortex. However, the components forming the SVZ stem cell niche in the developing neocortex have not yet been fully understood. In this review, we will discuss potential components of the SVZ stem cell niche, i.e., extracellular matrix composition and brain vasculature, and their possible key role in establishing and maintaining this niche during fetal neocortical development. We will also emphasize the potential role of basal progenitor morphology in maintaining their proliferative capacity within the stem cell niche of the SVZ. Finally, we will focus on the use of brain organoids to i) understand the unique features of basal progenitors, notably basal radial glia; ii) study components of the SVZ stem cell niche; and iii) provide future directions on how to improve brain organoids, notably the organoid SVZ, and make them more reliable models of human neocortical development and evolution studies.
Collapse
Affiliation(s)
| | - Michael Heide
- Research Group Brain Development and Evolution, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| |
Collapse
|
14
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [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: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
15
|
Di Marco B, Vázquez-Marín J, Monyer H, Centanin L, Alfonso J. Spatial transcriptomics map of the embryonic mouse brain - a tool to explore neurogenesis. Biol Open 2023; 12:bio060151. [PMID: 37855382 PMCID: PMC10602001 DOI: 10.1242/bio.060151] [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: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
The developing brain has a well-organized anatomical structure comprising different types of neural and non-neural cells. Stem cells, progenitors and newborn neurons tightly interact with their neighbouring cells and tissue microenvironment, and this intricate interplay ultimately shapes the output of neurogenesis. Given the relevance of spatial cues during brain development, we acknowledge the necessity for a spatial transcriptomics map accessible to the neurodevelopmental community. To fulfil this need, we generated spatially resolved RNA sequencing (RNAseq) data from embryonic day 13.5 mouse brain sections immunostained for mitotic active neural and vascular cells. Unsupervised clustering defined specific cell type populations of diverse lineages and differentiation states. Differential expression analysis revealed unique transcriptional signatures across specific brain areas, uncovering novel features inherent to particular anatomical domains. Finally, we integrated existing single-cell RNAseq datasets into our spatial transcriptomics map, adding tissue context to single-cell RNAseq data. In summary, we provide a valuable tool that enables the exploration and discovery of unforeseen molecular players involved in neurogenesis, particularly in the crosstalk between different cell types.
Collapse
Affiliation(s)
- Barbara Di Marco
- Department of Clinical Neurobiology at the German Cancer Research Center (DKFZ) and the Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Javier Vázquez-Marín
- Center for Organismal Studies, Heidelberg University, Im Neuenheimer Feld 230, Heidelberg 69120, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology at the German Cancer Research Center (DKFZ) and the Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Lázaro Centanin
- Center for Organismal Studies, Heidelberg University, Im Neuenheimer Feld 230, Heidelberg 69120, Germany
| | - Julieta Alfonso
- Department of Clinical Neurobiology at the German Cancer Research Center (DKFZ) and the Medical Faculty of the Heidelberg University, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Crouch EE, Joseph T, Marsan E, Huang EJ. Disentangling brain vasculature in neurogenesis and neurodegeneration using single-cell transcriptomics. Trends Neurosci 2023; 46:551-565. [PMID: 37210315 PMCID: PMC10560453 DOI: 10.1016/j.tins.2023.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/15/2023] [Accepted: 04/26/2023] [Indexed: 05/22/2023]
Abstract
The vasculature is increasingly recognized to impact brain function in health and disease across the life span. During embryonic brain development, angiogenesis and neurogenesis are tightly coupled, coordinating the proliferation, differentiation, and migration of neural and glial progenitors. In the adult brain, neurovascular interactions continue to play essential roles in maintaining brain function and homeostasis. This review focuses on recent advances that leverage single-cell transcriptomics of vascular cells to uncover their subtypes, their organization and zonation in the embryonic and adult brain, and how dysfunction in neurovascular and gliovascular interactions contributes to the pathogenesis of neurodegenerative diseases. Finally, we highlight key challenges for future research in neurovascular biology.
Collapse
Affiliation(s)
- Elizabeth E Crouch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Tara Joseph
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Elise Marsan
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric J Huang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA; Pathology Service (113B), San Francisco Veterans Administration Health Care System, San Francisco, CA 94121, USA.
| |
Collapse
|
17
|
Bishop D, Schwarz Q, Wiszniak S. Endothelial-derived angiocrine factors as instructors of embryonic development. Front Cell Dev Biol 2023; 11:1172114. [PMID: 37457293 PMCID: PMC10339107 DOI: 10.3389/fcell.2023.1172114] [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: 02/23/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Blood vessels are well-known to play roles in organ development and repair, primarily owing to their fundamental function in delivering oxygen and nutrients to tissues to promote their growth and homeostasis. Endothelial cells however are not merely passive conduits for carrying blood. There is now evidence that endothelial cells of the vasculature actively regulate tissue-specific development, morphogenesis and organ function, as well as playing roles in disease and cancer. Angiocrine factors are growth factors, cytokines, signaling molecules or other regulators produced directly from endothelial cells to instruct a diverse range of signaling outcomes in the cellular microenvironment, and are critical mediators of the vascular control of organ function. The roles of angiocrine signaling are only beginning to be uncovered in diverse fields such as homeostasis, regeneration, organogenesis, stem-cell maintenance, cell differentiation and tumour growth. While in some cases the specific angiocrine factor involved in these processes has been identified, in many cases the molecular identity of the angiocrine factor(s) remain to be discovered, even though the importance of angiocrine signaling has been implicated. In this review, we will specifically focus on roles for endothelial-derived angiocrine signaling in instructing tissue morphogenesis and organogenesis during embryonic and perinatal development.
Collapse
|
18
|
Pombero A, Garcia-Lopez R, Martínez S. Pericyte-Glioblastoma Cell Interaction: A Key Target to Prevent Glioblastoma Progression. Cells 2023; 12:1324. [PMID: 37174724 PMCID: PMC10177553 DOI: 10.3390/cells12091324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Multiple biological processes rely on direct intercellular interactions to regulate cell proliferation and migration in embryonic development and cancer processes. Tumor development and growth depends on close interactions between cancer cells and cells in the tumor microenvironment. During embryonic development, morphogenetic signals and direct cell contacts control cell proliferation, polarity, and morphogenesis. Cancer cells communicate with cells in the tumor niche through molecular signals and intercellular contacts, thereby modifying the vascular architecture and antitumor surveillance processes and consequently enabling tumor growth and survival. While looking for cell-to-cell signaling mechanisms that are common to both brain development and cancer progression, we have studied the infiltration process in glioblastoma multiforme (GBM), which is the most malignant primary brain tumor and with the worst prognosis. Cell-to-cell contacts, by means of filopodia-like structures, between GBM cells and brain pericytes (PCs) are necessary for adequate cell signaling during cancer infiltration; similarly, contacts between embryonic regions, via cytonemes, are required for embryo regionalization and development. This GBM-PC interaction provokes two important changes in the physiological function of these perivascular cells, namely, (i) vascular co-option with changes in cell contractility and vascular malformation, and (ii) changes in the PC transcriptome, modifying the microvesicles and protein secretome, which leads to the development of an immunosuppressive phenotype that promotes tumor immune tolerance. Moreover, the GTPase Cdc42 regulates cell polarity across organisms, from yeast to humans, playing a central role in GBM cell-PC interaction and maintaining vascular co-option. As such, a review of the molecular and cellular mechanisms underlying the development and maintenance of the physical interactions between cancer cells and PCs is of particular interest.
Collapse
Affiliation(s)
- Ana Pombero
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (A.P.); (R.G.-L.)
| | - Raquel Garcia-Lopez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (A.P.); (R.G.-L.)
| | - Salvador Martínez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (A.P.); (R.G.-L.)
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-ISCIII, 46010 Valencia, Spain
| |
Collapse
|
19
|
Wit CB, Hiesinger PR. Neuronal filopodia: From stochastic dynamics to robustness of brain morphogenesis. Semin Cell Dev Biol 2023; 133:10-19. [PMID: 35397971 DOI: 10.1016/j.semcdb.2022.03.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/30/2022]
Abstract
Brain development relies on dynamic morphogenesis and interactions of neurons. Filopodia are thin and highly dynamic membrane protrusions that are critically required for neuronal development and neuronal interactions with the environment. Filopodial interactions are typically characterized by non-deterministic dynamics, yet their involvement in developmental processes leads to stereotypic and robust outcomes. Here, we discuss recent advances in our understanding of how filopodial dynamics contribute to neuronal differentiation, migration, axonal and dendritic growth and synapse formation. Many of these advances are brought about by improved methods of live observation in intact developing brains. Recent findings integrate known and novel roles ranging from exploratory sensors and decision-making agents to pools for selection and mechanical functions. Different types of filopodial dynamics thereby reveal non-deterministic subcellular decision-making processes as part of genetically encoded brain development.
Collapse
Affiliation(s)
- Charlotte B Wit
- Devision of Neurobiology, Institute of Biology, Freie Universität Berlin, Berlin, Germany
| | - P Robin Hiesinger
- Devision of Neurobiology, Institute of Biology, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Ishihara K, Takata K, Mizutani KI. Involvement of an Aberrant Vascular System in Neurodevelopmental, Neuropsychiatric, and Neuro-Degenerative Diseases. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010221. [PMID: 36676170 PMCID: PMC9866034 DOI: 10.3390/life13010221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The vascular system of the prenatal brain is crucial for the development of the central nervous system. Communication between vessels and neural cells is bidirectional, and dysfunctional communication can lead to neurodevelopmental diseases. In the present review, we introduce neurodevelopmental and neuropsychiatric diseases potentially caused by disturbances in the neurovascular system and discuss candidate genes responsible for neurovascular system impairments. In contrast to diseases that can manifest during the developing stage, we have also summarized the disturbances of the neurovascular system in neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. Furthermore, we discussed the role of abnormal vascularization and dysfunctional vessels in the development of neurovascular-related diseases.
Collapse
Affiliation(s)
- Keiichi Ishihara
- Department of Pathological Biochemistry, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Correspondence: ; Tel.: +81-75-595-4656
| | - Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Ken-ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| |
Collapse
|
21
|
Estrada-Meza C, Torres-Copado A, Loreti González-Melgoza L, Ruiz-Manriquez LM, De Donato M, Sharma A, Pathak S, Banerjee A, Paul S. Recent insights into the microRNA and long non-coding RNA-mediated regulation of stem cell populations. 3 Biotech 2022; 12:270. [PMID: 36101546 PMCID: PMC9464284 DOI: 10.1007/s13205-022-03343-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/29/2022] [Indexed: 12/19/2022] Open
Abstract
Stem cells are undifferentiated cells that have multi-lineage differentiation. The transition from self-renewal to differentiation requires rapid and extensive gene expression alterations. Since different stem cells exhibit diverse non-coding RNAs (ncRNAs) expression profiles, the critical roles of ncRNAs in stem cell reprogramming, pluripotency maintenance, and differentiation have been widely investigated over the past few years. Hence, in this current review, the two main categories of ncRNAs, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are discussed. While the primary way by which miRNAs restrict mRNA transcription is through miRNA-mRNA interaction, lncRNAs have a wide range of effects on mRNA functioning, including interactions with miRNAs. Both of these ncRNAs participate in the post-transcriptional regulation of crucial biological mechanisms, such as cell cycle regulation, apoptosis, aging, and cell fate decisions. These findings shed light on a previously unknown aspect of gene regulation in stem cell fate determination and behavior. Overall, we summarized the key roles of miRNAs (including exosomal miRNAs) and lncRNAs in the regulation of stem cell populations, such as cardiac, hematopoietic, mesenchymal, neural, and spermatogonial, as well ncRNAs' influence on malignancy through modulating cancer stem cells, which might significantly contribute to clinical stem cell therapy and in regenerative medicine.
Collapse
Affiliation(s)
- Carolina Estrada-Meza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Andrea Torres-Copado
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Luisa Loreti González-Melgoza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Marcos De Donato
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| |
Collapse
|
22
|
Crouch EE, Bhaduri A, Andrews MG, Cebrian-Silla A, Diafos LN, Birrueta JO, Wedderburn-Pugh K, Valenzuela EJ, Bennett NK, Eze UC, Sandoval-Espinosa C, Chen J, Mora C, Ross JM, Howard CE, Gonzalez-Granero S, Lozano JF, Vento M, Haeussler M, Paredes MF, Nakamura K, Garcia-Verdugo JM, Alvarez-Buylla A, Kriegstein AR, Huang EJ. Ensembles of endothelial and mural cells promote angiogenesis in prenatal human brain. Cell 2022; 185:3753-3769.e18. [PMID: 36179668 PMCID: PMC9550196 DOI: 10.1016/j.cell.2022.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/17/2022] [Accepted: 08/29/2022] [Indexed: 01/26/2023]
Abstract
Interactions between angiogenesis and neurogenesis regulate embryonic brain development. However, a comprehensive understanding of the stages of vascular cell maturation is lacking, especially in the prenatal human brain. Using fluorescence-activated cell sorting, single-cell transcriptomics, and histological and ultrastructural analyses, we show that an ensemble of endothelial and mural cell subtypes tile the brain vasculature during the second trimester. These vascular cells follow distinct developmental trajectories and utilize diverse signaling mechanisms, including collagen, laminin, and midkine, to facilitate cell-cell communication and maturation. Interestingly, our results reveal that tip cells, a subtype of endothelial cells, are highly enriched near the ventricular zone, the site of active neurogenesis. Consistent with these observations, prenatal vascular cells transplanted into cortical organoids exhibit restricted lineage potential that favors tip cells, promotes neurogenesis, and reduces cellular stress. Together, our results uncover important mechanisms into vascular maturation during this critical period of human brain development.
Collapse
Affiliation(s)
- Elizabeth E Crouch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Madeline G Andrews
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arantxa Cebrian-Silla
- The 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
| | - Loukas N Diafos
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Janeth Ochoa Birrueta
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kaylee Wedderburn-Pugh
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Edward J Valenzuela
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Neal K Bennett
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ugomma C Eze
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carmen Sandoval-Espinosa
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jiapei Chen
- Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cristina Mora
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jayden M Ross
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Clare E Howard
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Susana Gonzalez-Granero
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y 21 Biología Evolutiva, Universitat de València - Centro de Investigación Biomédica en Red 22 sobre Enfermedades Neurodegenerativas, Valencia, Spain
| | - Jaime Ferrer Lozano
- Department of Pathology, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Maximo Vento
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain; Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Maximilian Haeussler
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95060, USA
| | - Mercedes F Paredes
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ken Nakamura
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y 21 Biología Evolutiva, Universitat de València - Centro de Investigación Biomédica en Red 22 sobre Enfermedades Neurodegenerativas, Valencia, Spain
| | - Arturo Alvarez-Buylla
- The 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; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Arnold R Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric J Huang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Science Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Developmental & Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA; Pathology Service 113B, San Francisco Veterans Affairs Healthcare System, San Francisco, CA 94121, USA.
| |
Collapse
|
23
|
The neural stem cell secretome across neurodevelopment. Exp Neurol 2022; 355:114142. [PMID: 35709983 DOI: 10.1016/j.expneurol.2022.114142] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022]
Abstract
Neural stem cell (NSC) based therapies are at the forefront of regenerative medicine strategies to combat illness and injury of the central nervous system (CNS). In addition to their ability to produce new cells, NSCs secrete a variety of products, known collectively as the NSC secretome, that have been shown to ameliorate CNS disease pathology and promote recovery. As pre-clinical and clinical research to harness the NSC secretome for therapeutic purposes advances, a more thorough understanding of the endogenous NSC secretome can provide useful insight into the functional capabilities of NSCs. In this review, we focus on research investigating the autocrine and paracrine functions of the endogenous NSC secretome across life. Throughout development and adulthood, we find evidence that the NSC secretome is a critical component of how endogenous NSCs regulate themselves and their niche. We also find gaps in current literature, most notably in the clinically-relevant domain of endogenous NSC paracrine function in the injured CNS. Future investigations to further define the endogenous NSC secretome and its role in CNS tissue regulation are necessary to bolster our understanding of NSC-niche interactions and to aid in the generation of safe and effective NSC-based therapies.
Collapse
|
24
|
Vogenstahl J, Parrilla M, Acker-Palmer A, Segarra M. Vascular Regulation of Developmental Neurogenesis. Front Cell Dev Biol 2022; 10:890852. [PMID: 35573692 PMCID: PMC9099230 DOI: 10.3389/fcell.2022.890852] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Evolutionary studies indicate that the nervous system evolved prior to the vascular system, but the increasing complexity of organisms prompted the vascular system to emerge in order to meet the growing demand for oxygen and nutrient supply. In recent years, it has become apparent that the symbiotic communication between the nervous and the vascular systems goes beyond the exclusive covering of the demands on nutrients and oxygen carried by blood vessels. Indeed, this active interplay between both systems is crucial during the development of the central nervous system (CNS). Several neural-derived signals that initiate and regulate the vascularization of the CNS have been described, however less is known about the vascular signals that orchestrate the development of the CNS cytoarchitecture. Here, we focus on reviewing the effects of blood vessels in the process of neurogenesis during CNS development in vertebrates. In mammals, we describe the spatiotemporal features of vascular-driven neurogenesis in two brain regions that exhibit different neurogenic complexity in their germinal zone, the hindbrain and the forebrain.
Collapse
Affiliation(s)
- Johanna Vogenstahl
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Marta Parrilla
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
- *Correspondence: Amparo Acker-Palmer, ; Marta Segarra,
| | - Marta Segarra
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
- *Correspondence: Amparo Acker-Palmer, ; Marta Segarra,
| |
Collapse
|
25
|
Lin X, Chen L, Jullienne A, Zhang H, Salehi A, Hamer M, C. Holmes T, Obenaus A, Xu X. Longitudinal dynamics of microvascular recovery after acquired cortical injury. Acta Neuropathol Commun 2022; 10:59. [PMID: 35468870 PMCID: PMC9036719 DOI: 10.1186/s40478-022-01361-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023] Open
Abstract
Acquired brain injuries due to trauma damage the cortical vasculature, which in turn impairs blood flow to injured tissues. There are reports of vascular morphological recovery following traumatic brain injury, but the remodeling process has not been examined longitudinally in detail after injury in vivo. Understanding the dynamic processes that influence recovery is thus critically important. We evaluated the longitudinal and dynamic microvascular recovery and remodeling up to 2 months post injury using live brain miniscope and 2-photon microscopic imaging. The new imaging approaches captured dynamic morphological and functional recovery processes at high spatial and temporal resolution in vivo. Vessel painting documented the initial loss and subsequent temporal morphological vascular recovery at the injury site. Miniscopes were used to longitudinally image the temporal dynamics of vascular repair in vivo after brain injury in individual mice across each cohort. We observe near-immediate nascent growth of new vessels in and adjacent to the injury site that peaks between 14 and 21 days post injury. 2-photon microscopy confirms new vascular growth and further demonstrates differences between cortical layers after cortical injury: large vessels persist in deeper cortical layers (> 200 μm), while superficial layers exhibit a dense plexus of fine (and often non-perfused) vessels displaying regrowth. Functionally, blood flow increases mirror increasing vascular density. Filopodia development and endothelial sprouting is measurable within 3 days post injury that rapidly transforms regions devoid of vessels to dense vascular plexus in which new vessels become increasingly perfused. Within 7 days post injury, blood flow is observed in these nascent vessels. Behavioral analysis reveals improved vascular modulation after 9 days post injury, consistent with vascular regrowth. We conclude that morphological recovery events are closely linked to functional recovery of blood flow to the compromised tissues, which subsequently leads to improved behavioral outcomes.
Collapse
|
26
|
Qi J, Rittershaus A, Priya R, Mansingh S, Stainier DYR, Helker CSM. Apelin signaling dependent endocardial protrusions promote cardiac trabeculation in zebrafish. eLife 2022; 11:e73231. [PMID: 35225788 PMCID: PMC8916774 DOI: 10.7554/elife.73231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
During cardiac development, endocardial cells (EdCs) produce growth factors to promote myocardial morphogenesis and growth. In particular, EdCs produce neuregulin which is required for ventricular cardiomyocytes (CMs) to seed the multicellular ridges known as trabeculae. Defects in neuregulin signaling, or in endocardial sprouting toward CMs, cause hypotrabeculation. However, the mechanisms underlying endocardial sprouting remain largely unknown. Here, we first show by live imaging in zebrafish embryos that EdCs interact with CMs via dynamic membrane protrusions. After touching CMs, these protrusions remain in close contact with their target despite the vigorous cardiac contractions. Loss of the CM-derived peptide Apelin, or of the Apelin receptor, which is expressed in EdCs, leads to reduced endocardial sprouting and hypotrabeculation. Mechanistically, neuregulin signaling requires endocardial protrusions to induce extracellular signal-regulated kinase (Erk) activity in CMs and trigger their delamination. Altogether, these data show that Apelin signaling-dependent endocardial protrusions modulate CM behavior during trabeculation.
Collapse
Affiliation(s)
- Jialing Qi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Annegret Rittershaus
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Rashmi Priya
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Shivani Mansingh
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier YR Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Christian SM Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
27
|
Regulation of the Brain Neural Niche by Soluble Molecule Akhirin. J Dev Biol 2021; 9:jdb9030029. [PMID: 34449638 PMCID: PMC8395899 DOI: 10.3390/jdb9030029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 01/22/2023] Open
Abstract
In the central nervous system (CNS), which comprises the eyes, spinal cord, and brain, neural cells are produced by the repeated division of neural stem cells (NSCs) during the development of the CNS. Contrary to the notion that the CNS is relatively static with a limited cell turnover, cells with stem cell-like properties have been isolated from most neural tissues. The microenvironment, also known as the NSC niche, consists of NSCs/neural progenitor cells, other neurons, glial cells, and blood vessels; this niche is thought to regulate neurogenesis and the differentiation of NSCs into neurons and glia. Although it has been established that neurons, glia, and blood vessels interact with each other in a complex manner to generate neural tissues in the NSC niche, the underlying molecular mechanisms in the CNS niche are unclear. Herein, we would like to introduce the extracellular secreted protein, Akhirin (AKH; Akhi is the Bengali translation for eye). AKH is specifically expressed in the CNS niche-the ciliary body epithelium in the retina, the central canal of the spinal cord, the subventricular zone, and the subgranular zone of the dentate gyrus of the hippocampus-and is supposedly involved in NSC niche regulation. In this review, we discuss the role of AKH as a niche molecule during mouse brain formation.
Collapse
|
28
|
Peguera B, Segarra M, Acker-Palmer A. Neurovascular crosstalk coordinates the central nervous system development. Curr Opin Neurobiol 2021; 69:202-213. [PMID: 34077852 PMCID: PMC8411665 DOI: 10.1016/j.conb.2021.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022]
Abstract
Purpose of the review: The synchronic development of vascular and nervous systems is orchestrated by common molecules that regulate the communication between both systems. The identification of these common guiding cues and the developmental processes regulated by neurovascular communication are slowly emerging. In this review, we describe the molecules modulating the neurovascular development and their impact in processes such as angiogenesis, neurogenesis, neuronal migration, and brain homeostasis. Recent findings: Blood vessels not only are involved in nutrient and oxygen supply of the central nervous system (CNS) but also exert instrumental functions controlling developmental neurogenesis, CNS cytoarchitecture, and neuronal plasticity. Conversely, neurons modulate CNS vascularization and brain endothelial properties such as blood–brain barrier and vascular hyperemia. Summary: The integration of the active role of endothelial cells in the development and maintenance of neuronal function is important to obtain a more holistic view of the CNS complexity and also to understand how the vasculature is involved in neuropathological conditions.
Collapse
Affiliation(s)
- Blanca Peguera
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Marta Segarra
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, Frankfurt am Main, D-60438, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, Frankfurt am Main, D-60438, Germany; Max Planck Institute for Brain Research, Max-von-Laue-Str. 4 Frankfurt am Main, 60438, Germany.
| |
Collapse
|
29
|
Silva TP, Sousa-Luís R, Fernandes TG, Bekman EP, Rodrigues CAV, Vaz SH, Moreira LM, Hashimura Y, Jung S, Lee B, Carmo-Fonseca M, Cabral JMS. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol Bioeng 2021; 118:2781-2803. [PMID: 33871054 DOI: 10.1002/bit.27797] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Human-induced pluripotent stem cells (iPSCs) have great potential for disease modeling. However, generating iPSC-derived models to study brain diseases remains a challenge. In particular, the ability to recapitulate cerebellar development in vitro is still limited. We presented a reproducible and scalable production of cerebellar organoids by using the novel single-use Vertical-Wheel bioreactors, in which functional cerebellar neurons were obtained. Here, we evaluate the global gene expression profiles by RNA sequencing (RNA-seq) across cerebellar differentiation, demonstrating a faster cerebellar commitment in this novel dynamic differentiation protocol. Furthermore, transcriptomic profiles suggest a significant enrichment of extracellular matrix (ECM) in dynamic-derived cerebellar organoids, which can better mimic the neural microenvironment and support a consistent neuronal network. Thus, an efficient generation of organoids with cerebellar identity was achieved for the first time in a continuous process using a dynamic system without the need of organoids encapsulation in ECM-based hydrogels, allowing the possibility of large-scale production and application in high-throughput processes. The presence of factors that favors angiogenesis onset was also detected in dynamic conditions, which can enhance functional maturation of cerebellar organoids. We anticipate that large-scale production of cerebellar organoids may help developing models for drug screening, toxicological tests, and studying pathological pathways involved in cerebellar degeneration.
Collapse
Affiliation(s)
- Teresa P Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal, Portugal
| | - Leonilde M Moreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Brian Lee
- PBS Biotech, Camarillo, California, USA
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|