1
|
Tang Z, Li R, Guo X, Wang Z, Wu J. Regulation of blood-brain barrier integrity by brain microvascular endothelial cells in ischemic stroke: A therapeutic opportunity. Eur J Pharmacol 2025; 996:177553. [PMID: 40147580 DOI: 10.1016/j.ejphar.2025.177553] [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: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Stroke is the second leading cause of death from cardiovascular diseases. Brain microvascular endothelial cells (BMECs) are crucial in the treatment of cerebral ischemic stroke, as their functional status directly affects the integrity of the blood-brain barrier (BBB). This review systematically discusses the central role of BMECs in ischemia. The mitochondrial dysfunction and activation of apoptosis/necrosis pathways in BMECs directly disrupt the integrity of the BBB and the degradation of junctional complexes (such as TJs and AJs) further exacerbates its permeability. In the neurovascular unit (NVU), astrocytes, microglia, and pericytes regulate the function of BMECs by secreting cytokines (such as TGF-β and VEGF), showing dual effects of promoting repair and damage. The dynamic changes of transporters, including those from the ATP-binding cassette and solute carrier families, as well as ion channels and exchangers, such as potassium and calcium channels, offer novel insights for the development of targeted drug delivery systems.
Collapse
Affiliation(s)
- Ziqi Tang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| | - Ruoxi Li
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Xi Guo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China.
| |
Collapse
|
2
|
Waxman S, Villafranca-Baughman D, Phillippi J, Jakobs TC, Alarcon-Martinez L, Di Polo A, Sigal IA. Pericytes in the Optic Nerve Head. Prog Retin Eye Res 2025:101375. [PMID: 40449651 DOI: 10.1016/j.preteyeres.2025.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 05/28/2025] [Accepted: 05/29/2025] [Indexed: 06/03/2025]
Abstract
Pericytes are a unique population of contractile mural cells and an essential part of the microvasculature. In the retina and brain, pericytes play crucial roles in regulating blood flow, maintaining the blood-brain barrier, signaling with neighboring cells, and depositing extracellular matrix. Pericyte dysfunction is an early process in a variety of neurodegenerative conditions. However, remarkably little is known about pericytes at an early site of neurodegeneration in glaucoma, the optic nerve head (ONH). This work summarizes the current understanding of pericyte contributions to ONH physiology, identifies potential roles in glaucomatous pathophysiology, and uncovers open questions at the intersection of these areas. We surveyed the literature to identify the roles of ONH pericytes in the context of health and glaucoma. Additionally, we probed for the presence of pericytes along microvasculature in mouse, nonhuman primate, and human donor ONH tissues. We identified an association between factors influencing ONH dysfunction in glaucoma and factors influencing pericyte dysfunction in other neurodegenerative conditions. Pericytes exist in the mouse, nonhuman primate, and human ONH, implicating their capacity for local function. ONH pericytes represent a promising but underexplored target for treating microvascular impairment in glaucoma. Investigating the contribution of pericytes in both healthy and disease states can help inform mechanisms of dysfunction in glaucomatous pathology, paving the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Julie Phillippi
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, University of Pittsburgh
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | - Luis Alarcon-Martinez
- Centre for Eye Research Australia, Department of Ophthalmology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Ian A Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh PA, USA.
| |
Collapse
|
3
|
Li W, Zhang Y, Zhu H, Su N, Sun R, Mao X, Yang Q, Yuan S. CAVIN3 deficiency promotes vascular normalization in ocular neovascular disease via ERK/JAG1 signaling pathway. JCI Insight 2025; 10:e187836. [PMID: 40337864 DOI: 10.1172/jci.insight.187836] [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: 10/10/2024] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Multiple members of the caveolae-associated protein (Cavin) family are implicated in angiogenesis. However, the specific role of CAVIN3 in pathological angiogenesis within the eye remains unclear. The present study demonstrated that CAVIN3 knockdown in endothelial cells (ECs) promoted vascular normalization in ocular pathological neovascularization. Elevated CAVIN3 expression was observed in the ECs of retinal pigment epithelium/choroid complexes from patients with neovascular age-related macular degeneration and fibrovascular membranes from patients with proliferative diabetic retinopathy. Additionally, upregulated Cavin3 expression was detected in laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) mouse models. In both OIR and CNV mice, Cavin3 knockdown inhibited pathological neovascularization. Cavin3 deficiency further disrupted EC proliferation and vascular sprouting, thereby promoting vascular normalization by partially restoring microenvironmental hypoxia and reestablishing pericyte-EC interactions. Mechanistically, we demonstrated that zinc finger E-box-binding homeobox 1 (ZEB1) regulated CAVIN3 transcription in ECs under hypoxic conditions. CAVIN3 deficiency modulated pathological vascularization by inhibiting ERK phosphorylation, which downregulated jagged 1 (JAG1) expression. Conclusively, this study elucidated the protective role of endothelial CAVIN3 deficiency in pathological neovascularization models, addressing a gap in understanding the regulatory role of Cavins in angiogenesis. These findings suggested a therapeutic direction for ocular neovascular diseases.
Collapse
Affiliation(s)
- Weiqi Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yeran Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjing Zhu
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Na Su
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruxu Sun
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Yang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Collado-Cuadrado M, Balmori-de la Puente A, Rodríguez-Escolar I, Infante González-Mohino E, Alarcón-Torrecillas C, Pericacho M, Morchón R. Glyceraldehyde 3-Phosphate Dehydrogenase and Galectin from Dirofilaria immitis Excretory/Secretory Antigens Activate Proangiogenic Pathway in In Vitro Vascular Endothelial Cell Model. Animals (Basel) 2025; 15:964. [PMID: 40218357 PMCID: PMC11987766 DOI: 10.3390/ani15070964] [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: 01/21/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Heartworm disease is caused by Dirofilaria immitis, which mainly affects canids and felids. Adult D. immitis worms are located between the heart's right ventricle and the pulmonary artery. These parasites produce an inflammatory and hypoxic process in the vascular endothelium. It has been demonstrated that D. immitis excretory/secretory antigens are able to stimulate the angiogenic process as a survival mechanism of D. immitis in the vascular endothelium, stimulating the proangiogenic pathway and related cellular processes. Our goal was to study the role of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and galectin (GAL) (proteins of D. immitis excretory/secretory antigens) plus vascular endothelial growth factor isoform A (VEGF-A) in the angiogenic process and their relationship with three cellular processes (cell proliferation, cell migration, and pseudocapillary formation) in an in vitro model of vascular endothelial cells. Cell viability and cytotoxicity were analyzed by live cell analysis and a commercial kit, respectively. VEGF-A, sVEGFR-2, VEGFR-1/sFlt, soluble endoglin, and membrane endoglin were analyzed by commercial ELISA kits. Cell proliferation, cell migration, and pseudocapillary formation were analyzed by MTT-based assay, the wound healing technique, and counting cell connections and cell clusters, respectively. rDiGAPDH+VEGF-A and rDiGAL+VEGF-A significantly increased the expression of sVEGFR-2, mEndoglin, and VEGF-A compared to cultures treated with only the proteins (rDiGAPDH and rDiGAL), VEGF-A, or unstimulated cultures. In addition, they also produced a significant increase in cell proliferation, cell migration, and pseudocapillary formation. Therefore, these proteins together with VEGF-A can activate the proangiogenic pathway and could be related to D. immitis survival in the circulatory system.
Collapse
Affiliation(s)
- Manuel Collado-Cuadrado
- Zoonotic Diseases and One Health Group, Faculty of Pharmacy, Biomedical Research Institute of Salamanca (IBSAL), Centre for Environmental Studies and Rural Dynamization (CEADIR), University of Salamanca, 37007 Salamanca, Spain; (M.C.-C.); (A.B.-d.l.P.); (I.R.-E.); (E.I.G.-M.)
| | - Alfonso Balmori-de la Puente
- Zoonotic Diseases and One Health Group, Faculty of Pharmacy, Biomedical Research Institute of Salamanca (IBSAL), Centre for Environmental Studies and Rural Dynamization (CEADIR), University of Salamanca, 37007 Salamanca, Spain; (M.C.-C.); (A.B.-d.l.P.); (I.R.-E.); (E.I.G.-M.)
| | - Iván Rodríguez-Escolar
- Zoonotic Diseases and One Health Group, Faculty of Pharmacy, Biomedical Research Institute of Salamanca (IBSAL), Centre for Environmental Studies and Rural Dynamization (CEADIR), University of Salamanca, 37007 Salamanca, Spain; (M.C.-C.); (A.B.-d.l.P.); (I.R.-E.); (E.I.G.-M.)
| | - Elena Infante González-Mohino
- Zoonotic Diseases and One Health Group, Faculty of Pharmacy, Biomedical Research Institute of Salamanca (IBSAL), Centre for Environmental Studies and Rural Dynamization (CEADIR), University of Salamanca, 37007 Salamanca, Spain; (M.C.-C.); (A.B.-d.l.P.); (I.R.-E.); (E.I.G.-M.)
| | - Claudia Alarcón-Torrecillas
- Department of Physiology and Pharmacology, Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain; (C.A.-T.); (M.P.)
| | - Miguel Pericacho
- Department of Physiology and Pharmacology, Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain; (C.A.-T.); (M.P.)
| | - Rodrigo Morchón
- Zoonotic Diseases and One Health Group, Faculty of Pharmacy, Biomedical Research Institute of Salamanca (IBSAL), Centre for Environmental Studies and Rural Dynamization (CEADIR), University of Salamanca, 37007 Salamanca, Spain; (M.C.-C.); (A.B.-d.l.P.); (I.R.-E.); (E.I.G.-M.)
| |
Collapse
|
5
|
Lin YY, Warren E, Macklin BL, Ramirez L, Gerecht S. Endothelial-pericyte interactions regulate angiogenesis via VEGFR2 signaling during retinal development and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642174. [PMID: 40161680 PMCID: PMC11952325 DOI: 10.1101/2025.03.08.642174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pericytes stabilize the microvasculature by enhancing endothelial barrier integrity, resulting in functional networks. During retinal development, pericyte recruitment is crucial for stabilizing nascent angiogenic vasculature. However, in adulthood, disrupted endothelial-pericyte interactions lead to vascular dropout and pathological angiogenesis in ocular microvascular diseases, and strategies to stabilize the retinal vasculature are lacking. We demonstrate that direct endothelial-pericyte contact downregulates pVEGFR2 in endothelial cells, which enhances pericyte migration and promotes endothelial cell barrier function. Intravitreal injection of a VEGFR2 inhibitor in mouse models of the developing retina and oxygen-induced retinopathy increased pericyte recruitment and aided vascular stability. The VEGFR2 inhibitor further rescued ischemic retinopathy by enhancing vascularization and tissue growth while reducing vascular permeability. Our findings offer a druggable target to support the growth of functional and mature microvasculature in ocular microvascular diseases and tissue regeneration overall.
Collapse
|
6
|
Atak D, Yıldız E, Özkan E, Yousefi M, Özkan A, Yılmaz AB, Kızılırmak AB, Alnajjar IA, Kanar Ç, Caan ZL, Zeybek ŞÜ, Küçükali Cİ, Tüzün E, Gürsoy‐Özdemir Y, Vural A. Longitudinal Investigation of Brain and Spinal Cord Pericytes After Inducible PDGFRβ + Cell Ablation in Adult Mice. J Neurochem 2025; 169:e70035. [PMID: 40066845 PMCID: PMC11894923 DOI: 10.1111/jnc.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
Central nervous system (CNS) pericytes play crucial roles in vascular development and blood-brain barrier maturation during prenatal development, as well as in regulating cerebral blood flow in adults. They have also been implicated in the pathogenesis of numerous neurological disorders. However, the behavior of pericytes in the adult brain after injury remains poorly understood, partly due to limitations in existing pericyte ablation models. To investigate pericyte responses following acute ablation and characterize a novel rodent model for pericyte research, we developed a tamoxifen-inducible PDGFRβ+ cell ablation model by crossing PDGFRβ-P2A-CreERT2 and Rosa26-DTA176 transgenic mouse lines. Using this model, we studied the effects of different tamoxifen doses and conducted histological examinations 15 and 60 days post-injection to assess the impacts of PDGFRβ+ cell ablation in both acute and chronic phases, respectively. Our results demonstrate that a low dose of tamoxifen effectively ablates PDGFRβ+ cells of the CNS in mice without reducing survival or causing significant systemic side effects, such as weight loss. Additionally, we found that the extent of PDGFRβ+ cell depletion varies between the cortex and the spinal cord, as well as between the gray and white matter regions of the spinal cord. Importantly, we observed that both pericyte coverage and numbers increased in the weeks following acute ablation, indicating the regenerative capacity of CNS pericytes in vivo. This study offers a valuable tool for future studies on the role of pericytes in neurological disorders by overcoming the limitations of constitutive pericyte ablation models and providing its longitudinal characterization in the CNS.
Collapse
Affiliation(s)
- Dila Atak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Erdost Yıldız
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Esra Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Mohammadreza Yousefi
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ayşe Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of PhysiologyBakırçay UniversityİzmirTürkiye
| | - Aysu Bilge Yılmaz
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ali Burak Kızılırmak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | | | - Çiçek Kanar
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Zeynep Lal Caan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Şakir Ümit Zeybek
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Cem İsmail Küçükali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Yasemin Gürsoy‐Özdemir
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Atay Vural
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| |
Collapse
|
7
|
Álvarez-Aznar A, Desai M, Orlich MM, Vázquez-Liébanas E, Adams RH, Brakebusch C, Gaengel K. Cdc42 is crucial for mural cell migration, proliferation and patterning of the retinal vasculature. Vascul Pharmacol 2025; 159:107472. [PMID: 39971261 DOI: 10.1016/j.vph.2025.107472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
AIMS Mural cells constitute the outer lining of blood vessels and are essential for vascular development and function. Mural cell loss or malfunction has been associated with numerous diseases including diabetic retinopathy, stroke and amyotrophic lateral sclerosis. In this work, we investigate the role of CDC42 in mural cells in vivo, using the developing mouse retina as a model. METHODS In this study, we generated a mouse model for Cdc42 deletion in mural cells by crossing Pdgfrb-CreERT2 mice with Cdc42flox/flox mice. This model (Cdc42iΔMC) allowed us to investigate the role of CDC42 in pericytes and smooth muscle cells in the developing and adult retinal vasculature. RESULTS We find that, during postnatal development, CDC42 is required in both, pericytes and smooth muscle cells to maintain proper cell morphology, mural cell coverage and distribution. During retinal angiogenesis, Cdc42-depleted pericytes lag behind the sprouting front and exhibit decreased proliferation. Consequently, capillaries at the sprouting front remain pericyte deprived, become dilated and are prone to increased vascular leakage. In addition, arteries and arterioles deviate from their normal growth directions and trajectory. While in the adult retina, mural cell coverage normalizes and pericytes adopt a normal morphology, smooth muscle cell morphologies remain abnormal and arteriolar branching angles are markedly reduced. CONCLUSIONS Our findings demonstrate that CDC42 is required for mural cell migration and proliferation and suggest that mural cells are essential for normal morphogenesis and patterning of the developing retinal vasculature.
Collapse
Affiliation(s)
- Alberto Álvarez-Aznar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Malavika Desai
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael M Orlich
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ralf H Adams
- Max Plank Institute for Molecular Medicine, Department of Tissue Morphogenesis, Münster, Germany
| | | | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Grootaert MOJ, Pasut A, Raman J, Simmonds SJ, Callewaert B, Col Ü, Dewerchin M, Carmeliet P, Heymans S, Jones EAV. Mural cell dysfunction contributes to diastolic heart failure by promoting endothelial dysfunction and vessel remodelling. Cardiovasc Diabetol 2025; 24:62. [PMID: 39920783 PMCID: PMC11806843 DOI: 10.1186/s12933-025-02623-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a complex cardiovascular disease associated with metabolic comorbidities. Microvascular dysfunction has been proposed to drive HFpEF, likely via endothelial cell (EC) dysfunction, yet the role of the mural cells herein has never been explored. METHODS We used the diabetic db/db mouse given 1% salt as a new model of HFpEF and crossed then with PDGFRβtg/tg-CreERT2-EYFPtg/tg mice to label the mural cells. We combined single-cell RNA sequencing, NichetNet analysis and histology to determine the role of mural cell dysfunction in HFpEF. RESULTS Db/db mice given 1% salt for 8 weeks developed diastolic dysfunction preceded by capillary density loss, pericyte loss and vessel regression. At 4 weeks of salt, hearts of db/db mice already showed EC dysfunction associated with an anti-angiogenic signature, and an increase in pericyte-EC intracellular space. Db/db + salt hearts were further characterised by increased ACTA2 expression, arteriole wall thickening and vessel enlargement. NicheNet analysis on the single cell transcriptomic data revealed little signalling from the ECs to the mural cells; instead, mural cells signalled strongly to ECs. Mechanistically, pericyte dysfunction induces an EC growth arrest via TNFα-dependent paracrine signalling and downstream signalling through STAT1. CONCLUSION Mural cell dysfunction contributes to HFpEF by inducing coronary vessel remodelling, at least in part by reducing EC proliferation and inducing EC inflammation through TNFα-dependent paracrine signalling.
Collapse
MESH Headings
- Animals
- Pericytes/metabolism
- Pericytes/pathology
- Disease Models, Animal
- Vascular Remodeling
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Heart Failure, Diastolic/physiopathology
- Heart Failure, Diastolic/metabolism
- Heart Failure, Diastolic/pathology
- Heart Failure, Diastolic/genetics
- Heart Failure, Diastolic/etiology
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Coronary Vessels/pathology
- Signal Transduction
- Ventricular Function, Left
- Male
- Mice, Transgenic
- Mice, Inbred C57BL
- Stroke Volume
- Mice
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Paracrine Communication
- Actins
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Endocrinology, Diabetes and Nutrition, UCLouvain, Avenue Hippocrate, box 55, Brussels, 1200, Belgium.
| | - Alessandra Pasut
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
| | - Jana Raman
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Steven J Simmonds
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Bram Callewaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Ümare Col
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| |
Collapse
|
9
|
Das UN. Lipoxin A4 (LXA4) as a Potential Drug for Diabetic Retinopathy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:177. [PMID: 40005295 PMCID: PMC11857424 DOI: 10.3390/medicina61020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025]
Abstract
The purpose of this review is to propose that lipoxin A4 (LXA4), derived from arachidonic acid (AA), a potent anti-inflammatory, cytoprotective, and wound healing agent, may be useful to prevent and manage diabetic retinopathy (DR). LXA4 suppresses inappropriate angiogenesis and the production of pro-inflammatory prostaglandin E2 (PGE2), leukotrienes (LTs), 12-HETE (12-hydroxyeicosatetraenoic acid), derived from AA by the action of 12-lioxygenase (12-LOX)) interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), as well as the expression of NF-κB, inducible NO (nitric oxide) synthase (iNOS), cyclooxygenase-2 (COX-2), intracellular adhesion molecule-1 (ICAM-1), and vascular endothelial growth factor (VEGF)-factors that play a role in DR. Thus, the intravitreal injection of LXA4 may form a new approach to the treatment of DR and other similar conditions such as AMD (age-associated macular degeneration) and SARS-CoV-2-associated hyperinflammatory immune response in the retina. The data for this review are derived from our previous work conducted in individuals with DR and from various publications on LXA4, inflammation, and DR.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA 98604, USA
| |
Collapse
|
10
|
Huang C, Zhang X, Wu M, Yang C, Ge X, Chen W, Li X, Liu S, Yang S. IL-1β-induced pericyte dysfunction with a secretory phenotype exacerbates retinal microenvironment inflammation via Hes1/STAT3 signaling pathway. Int Immunopharmacol 2025; 144:113611. [PMID: 39612772 DOI: 10.1016/j.intimp.2024.113611] [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: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024]
Abstract
Retinal pericytes are mural cells surrounding capillaries to maintain the integrity of blood-retina barrier and regulate vascular behaviors. Pericyte loss has been considered as the hallmark of diabetic retinopathy (DR), which is a major complication of diabetes and the leading cause of blindness in adults. However, the precise function of pericytes in regulating the retinal microenvironment and the underlying mechanism remains largely unknown. In this study, we observed a secretory phenotype of pericytes with elevated inflammatory cytokines in response to Interleukin-1β (IL-1β), a canonical inflammatory cytokine which significantly increases during the initial stages of diabetic retinopathy. This phenotype is also accompanied by reduced expression of adherent junction proteins and contractile proteins. Paracrine cytokines derived from pericytes further induce the chemotaxis of microglia cells and trigger detrimental changes in endothelial cells, including reduced expression of tight junction protein Occludin and increased apoptosis. Mechanically, the secretion potential in pericytes is partially mediated by Hes1/STAT3 signaling pathway. Moreover, co-injection of stattic, an inhibitor targeting STAT3 activation, could effectively attenuate IL-1β-induced retinal inflammation and microglial activation in retina tissues. Collectively, these findings demonstrate the potential of retinal pericytes as an initial inflammatory sensor prior to their anatomical pathological loss, via undergoing phenotypic changes and secreting paracrine factors to amplify local inflammation and damage endothelial cells in vitro. Furthermore, inhibition of STAT3 activation by inhibitors significantly ameliorates IL-1β-induced retinal inflammation, suggesting STAT3 in retinal pericytes as a promising target for alleviating DR and other IL-1β-induced ocular diseases.
Collapse
Affiliation(s)
- Caoxin Huang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Xiaofang Zhang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Menghua Wu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chen Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xilin Ge
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenting Chen
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xuejun Li
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Suhuan Liu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Shuyu Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
11
|
Morton L, Garza AP, Debska‐Vielhaber G, Villafuerte LE, Henneicke S, Arndt P, Meuth SG, Schreiber S, Dunay IR. Pericytes and Extracellular Vesicle Interactions in Neurovascular Adaptation to Chronic Arterial Hypertension. J Am Heart Assoc 2025; 14:e038457. [PMID: 39719419 PMCID: PMC12054408 DOI: 10.1161/jaha.124.038457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Chronic arterial hypertension restructures the vascular architecture of the brain, leading to a series of pathological responses that culminate in cerebral small-vessel disease. Pericytes respond dynamically to vascular challenges; however, how they manifest under the continuous strain of hypertension has not been elucidated. METHODS AND RESULTS In this study, we characterized pericyte behavior alongside hypertensive states in the spontaneously hypertensive stroke-prone rat model, focusing on their phenotypic and metabolic transformation. Flow cytometry was used to characterize pericytes by their expression of platelet-derived growth factor receptor β, neuroglial antigen 2, cluster of differentiation 13-alanyl aminopeptidase, and antigen Kiel 67. Microvessels were isolated for gene expression profiling and in vitro pericyte expansion. Immunofluorescence validated the cell culture model. Plasma-derived extracellular vesicles from hypertensive rodents were applied as a treatment to assess their effects on pericyte function and detailed metabolic assessments on enriched pericytes measured oxidative phosphorylation and glycolysis. Our results reveal a shift in platelet-derived growth factor receptor β+ pericytes toward increased neuroglial antigen 2 and cluster of differentiation 13-alanyl aminopeptidase coexpression, indicative of their critical role in vascular stabilization and inflammatory responses within the hypertensive milieu. Significant alterations were found within key pathways including angiogenesis, blood-brain barrier integrity, hypoxia, and inflammation. Circulating extracellular vesicles from hypertensive rodents distinctly influenced pericyte mitochondrial function, evidencing their dual role as carriers of disease pathology and potential therapeutic agents. Furthermore, a shift toward glycolytic metabolism in hypertensive pericytes was confirmed, coupled with ATP production dysregulation. CONCLUSIONS Our findings demonstrate that cerebral pericytes undergo phenotypic and metabolic reprogramming in response to hypertension, with hypertensive-derived plasma-derived extracellular vesicles impairing their mitochondrial function. Importantly, plasma-derived extracellular vesicles from normotensive controls restore this function, suggesting their potential as both therapeutic agents and precision biomarkers for hypertensive vascular complications. Further investigation into plasma-derived extracellular vesicle cargo is essential to further explore their therapeutic potential in vascular health.
Collapse
Affiliation(s)
- Lorena Morton
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | - Alejandra P. Garza
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | | | - Luis E. Villafuerte
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | - Solveig Henneicke
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
| | - Philipp Arndt
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
| | - Sven G. Meuth
- Department of NeurologyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Stefanie Schreiber
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
- Center for Behavioral Brain Sciences (CBBS)MagdeburgGermany
- German Center for Mental Health (DZPG)Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C‐I‐R‐C)Halle‐Jena‐MagdeburgGermany
| | - Ildiko R. Dunay
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
- Center for Behavioral Brain Sciences (CBBS)MagdeburgGermany
- German Center for Mental Health (DZPG)Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C‐I‐R‐C)Halle‐Jena‐MagdeburgGermany
| |
Collapse
|
12
|
Cashion JM, Brown LS, Morris GP, Fortune AJ, Courtney JM, Makowiecki K, Premilovac D, Cullen CL, Young KM, Sutherland BA. Pericyte ablation causes hypoactivity and reactive gliosis in adult mice. Brain Behav Immun 2025; 123:681-696. [PMID: 39406266 DOI: 10.1016/j.bbi.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024] Open
Abstract
Capillary pericytes are important regulators of cerebral blood flow, blood-brain barrier integrity and neuroinflammation, but can become lost or dysfunctional in disease. The consequences of pericyte loss or dysfunction is extremely difficult to discern when it forms one component of a complex disease process. To evaluate this directly, we examined the effect of adult pericyte loss on mouse voluntary movement and motor function, and physiological responses such as hypoxia, blood-brain barrier (BBB) integrity and glial reactivity. Tamoxifen delivery to Pdgfrβ-CreERT2:: Rosa26-DTA transgenic mice was titrated to produce a dose-dependent ablation of pericytes in vivo. 100mg/kg of tamoxifen ablated approximately half of all brain pericytes, while two consecutive daily doses of 300mg/kg tamoxifen ablated >80% of brain pericytes. In the open field test, mice with ∼50% pericyte loss spent more time immobile and travelled half the distance of control mice. Mice with >80% pericyte ablation also slipped more frequently while performing the beam walk task. Our histopathological analyses of the brain revealed that blood vessel density was unchanged, but vessel lumen width was increased. Pericyte-ablated mice also exhibited: mild BBB disruption; increased neuronal hypoxia; astrogliosis and increased IBA1+ immunoreactivity, suggestive of microgliosis and/or macrophage infiltration. Our results highlight the importance of pericytes in the brain, as pericyte loss can directly compromise brain health and induce behavioural alterations in mice.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Alastair J Fortune
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jo-Maree Courtney
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Dino Premilovac
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
13
|
Salazar-Saura I, Pinilla-Sala M, Megías J, Navarro L, Roselló-Sastre E, San-Miguel T. Pericytes in Glioblastoma: Hidden Regulators of Tumor Vasculature and Therapy Resistance. Cancers (Basel) 2024; 17:15. [PMID: 39796646 PMCID: PMC11718950 DOI: 10.3390/cancers17010015] [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: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Glioblastoma IDH wild type (GB), the most common malignant primary brain tumor, is characterized by rapid proliferation, extensive infiltration into surrounding brain tissue, and significant resistance to current therapies. Median survival is only 15 months despite extensive clinical efforts. The tumor microenvironment (TME) in GB is highly specialized, supporting the tumor's aggressive behavior and its ability to evade conventional treatments. One critical component is the aberrant vascular network that complicates the delivery of chemotherapy across the blood-brain barrier. Antiangiogenic therapies emerged as a promising option but have shown limited efficacy in extending the survival of these patients. Comprehension of the complex vascular network of GB may be a key to overcoming the limitations of current therapies. Pericytes are gaining recognition within the context of the TME. These mural cells are essential for vascular integrity and may contribute to tumor progression and therapeutic resistance. Although their role has been evidenced in other tumors, they remain underexplored in GB. Pericytes are known to respond to tumor hypoxia and interact with vascular endothelia, influencing responses to DNA damage and antiangiogenic treatments. They actively regulate not only angiogenesis but also the different vasculogenic strategies for tumor neovascularization. Additionally, they affect leukocyte trafficking and tumor-associated macrophages. This review aims to integrate the various functions controlled by pericytes to favor deeper investigation into their actionable potential. Pericytes may represent a promising target for novel therapeutic strategies in order to improve patient outcomes.
Collapse
Affiliation(s)
- Irene Salazar-Saura
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - María Pinilla-Sala
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Javier Megías
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Lara Navarro
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Esther Roselló-Sastre
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Teresa San-Miguel
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| |
Collapse
|
14
|
Sun WJ, An XD, Zhang YH, Tang SS, Sun YT, Kang XM, Jiang LL, Zhao XF, Gao Q, Ji HY, Lian FM. Autophagy-dependent ferroptosis may play a critical role in early stages of diabetic retinopathy. World J Diabetes 2024; 15:2189-2202. [PMID: 39582563 PMCID: PMC11580571 DOI: 10.4239/wjd.v15.i11.2189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetic retinopathy (DR), as one of the most common and significant microvascular complications of diabetes mellitus (DM), continues to elude effective targeted treatment for vision loss despite ongoing enrichment of the understanding of its pathogenic mechanisms from perspectives such as inflammation and oxidative stress. Recent studies have indicated that characteristic neuroglial degeneration induced by DM occurs before the onset of apparent microvascular lesions. In order to comprehensively grasp the early-stage pathological changes of DR, the retinal neurovascular unit (NVU) will become a crucial focal point for future research into the occurrence and progression of DR. Based on existing evidence, ferroptosis, a form of cell death regulated by processes like ferritinophagy and chaperone-mediated autophagy, mediates apoptosis in retinal NVU components, including pericytes and ganglion cells. Autophagy-dependent ferroptosis-related factors, including BECN1 and FABP4, may serve as both biomarkers for DR occurrence and development and potentially crucial targets for future effective DR treatments. The aforementioned findings present novel perspectives for comprehending the mechanisms underlying the early-stage pathological alterations in DR and open up innovative avenues for investigating supplementary therapeutic strategies.
Collapse
Affiliation(s)
- Wen-Jie Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Dong An
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Yue-Hong Zhang
- Department of Endocrinology, Fangshan Hospital of Beijing University of Chinese Medicine, Beijing 102400, China
| | - Shan-Shan Tang
- Department of Endocrinology, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Yu-Ting Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xiao-Min Kang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Lin-Lin Jiang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Fei Zhao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Qing Gao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Hang-Yu Ji
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Feng-Mei Lian
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| |
Collapse
|
15
|
Xiao J, Xu Z. Roles of noncoding RNAs in diabetic retinopathy: Mechanisms and therapeutic implications. Life Sci 2024; 357:123092. [PMID: 39368772 DOI: 10.1016/j.lfs.2024.123092] [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: 07/09/2024] [Revised: 08/20/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
Diabetic retinopathy (DR) is a microvascular complication of diabetes that leads to vision loss. The striking features of DR are hard exudate, cotton-wool spots, hemorrhage, and neovascularization. The dysregulated retinal cells, encompassing microvascular endothelial cells, pericytes, Müller cells, and adjacent retinal pigment epithelial cells, are involved in the pathological processes of DR. According to recent research, oxidative stress, inflammation, ferroptosis, pyroptosis, apoptosis, and angiogenesis contribute to DR. Recent advancements have highlighted that noncoding RNAs could regulate diverse targets in pathological processes that contribute to DR. Noncoding RNAs, including long noncoding RNAs, microRNAs (miRNA), and circular RNAs, are dysregulated in DR, and interact with miRNA, mRNA, or proteins to control the pathological processes of DR. Hence, modulation of noncoding RNAs may have therapeutic effects on DR. Small extracellular vesicles may be valuable tools for transferring noncoding RNAs and regulating the genes involved in progression of DR. However, the roles of noncoding RNA in developing DR are not fully understood; it is critical to summarize the mechanisms for noncoding RNA regulation of pathological processes and pathways related to DR. This review provides a fundamental understanding of the relationship between noncoding RNAs and DR, exploring the mechanism of how noncoding RNA modulates different signaling pathways, and pave the way for finding potential therapeutic strategies for DR.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
16
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
17
|
Bannykh KS, Fuentes-Fayos AC, Linesch PW, Breunig JJ, Bannykh SI. Laminin Beta 2 Is Localized at the Sites of Blood-Brain Barrier and Its Disruption Is Associated With Increased Vascular Permeability, Histochemical, and Transcriptomic Study. J Histochem Cytochem 2024; 72:641-667. [PMID: 39340425 PMCID: PMC11472343 DOI: 10.1369/00221554241281896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Heterotrimeric extracellular matrix proteins laminins are mostly deposited at basal membranes and are important in repair and neoplasia. Here, we localize laminin beta 2 (LAMB2) at the sites of blood-brain barrier (BBB). Microvasculature (MV) of normal brain is endowed with complete LAMB2 coverage. In contrast, its cognate protein laminin beta 1 (LAMB1) is absent in MV of normal brain but emerges at the sprouting tip of a growing vessels. Similarly, vascular proliferation in high-grade gliomas (HGG) is accompanied by marked overexpression of LAMB1, whereas LAMB2 shows deficient deposition. We find that many brain pathologies with presence of post-gadolinium enhancement (PGE) on magnetic resonance imaging (MRI) show disruption of LAMB2 vascular ensheathment. Inhibition of vascular endothelial growth factor signaling in HGG blocks angiogenesis, suppresses PGE in HGG, prevents expression of LAMB1, and restores LAMB2 vascular coverage. Analysis of single-cell RNA sequencing (scRNA-seq) databases shows that in quiescent brain LAMB2 is predominantly expressed by BBB-associated pericytes (PCs) and endothelial cells (ECs), whereas neither cell types produce LAMB1. In contrast, in HGG, both LAMB1 and 2 are overexpressed by endothelial precursor cells, a phenotypically unique immature group, specific to proliferating hyperplastic MV.
Collapse
Affiliation(s)
- Katherine S. Bannykh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Antonio C. Fuentes-Fayos
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Paul W. Linesch
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J. Breunig
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Serguei I. Bannykh
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
18
|
Tang Y, Frisendahl C, Piltonen TT, Arffman RK, Lalitkumar PG, Gemzell-Danielsson K. Human Endometrial Pericytes: A Comprehensive Overview of Their Physiological Functions and Implications in Uterine Disorders. Cells 2024; 13:1510. [PMID: 39273080 PMCID: PMC11394273 DOI: 10.3390/cells13171510] [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: 07/25/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Pericytes are versatile cells integral to the blood vessel walls of the microcirculation, where they exhibit specific stem cell traits. They are essential in modulating blood flow, ensuring vascular permeability, and maintaining homeostasis and are involved in the tissue repair process. The human endometrium is a unique and complex tissue that serves as a natural scar-free healing model with its cyclical repair and regeneration process every month. The regulation of pericytes has gained increasing attention due to their involvement in various physiological and pathological processes. However, endometrial pericytes are less well studied compared to the pericytes in other organs. This review aims to provide a comprehensive overview of endometrial pericytes, with a focus on elucidating their physiological function and potential implications in uterine disorders.
Collapse
Affiliation(s)
- Yiqun Tang
- WHO Collaborating Centre, Division of Neonatology, Obstetrics, Gynecology, and Reproductive Health, Department of Women’s and Children’s Health, Karolinska University Hospital, Karolinska Institutet, SE 17176 Stockholm, Sweden; (Y.T.); (C.F.); (P.G.L.)
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Centre, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland; (T.T.P.); (R.K.A.)
| | - Caroline Frisendahl
- WHO Collaborating Centre, Division of Neonatology, Obstetrics, Gynecology, and Reproductive Health, Department of Women’s and Children’s Health, Karolinska University Hospital, Karolinska Institutet, SE 17176 Stockholm, Sweden; (Y.T.); (C.F.); (P.G.L.)
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Centre, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland; (T.T.P.); (R.K.A.)
| | - Riikka K. Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Centre, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland; (T.T.P.); (R.K.A.)
| | - Parameswaran Grace Lalitkumar
- WHO Collaborating Centre, Division of Neonatology, Obstetrics, Gynecology, and Reproductive Health, Department of Women’s and Children’s Health, Karolinska University Hospital, Karolinska Institutet, SE 17176 Stockholm, Sweden; (Y.T.); (C.F.); (P.G.L.)
| | - Kristina Gemzell-Danielsson
- WHO Collaborating Centre, Division of Neonatology, Obstetrics, Gynecology, and Reproductive Health, Department of Women’s and Children’s Health, Karolinska University Hospital, Karolinska Institutet, SE 17176 Stockholm, Sweden; (Y.T.); (C.F.); (P.G.L.)
| |
Collapse
|
19
|
Liu B, Zeng H, Su H, Williams QA, Besanson J, Chen Y, Chen J. Endothelial Cell-Specific Prolyl Hydroxylase-2 Deficiency Augments Angiotensin II-Induced Arterial Stiffness and Cardiac Pericyte Recruitment in Mice. J Am Heart Assoc 2024; 13:e035769. [PMID: 39056332 PMCID: PMC11963933 DOI: 10.1161/jaha.124.035769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Endothelial prolyl hydroxylase-2 (PHD2) is essential for pulmonary remodeling and hypertension. In the present study, we investigated the role of endothelial PHD2 in angiotensin II-mediated arterial stiffness, pericyte recruitment, and cardiac fibrosis. METHODS AND RESULTS Chondroitin sulfate proteoglycan 4 tracing reporter chondroitin sulfate proteoglycan 4- red fluorescent protein (DsRed) transgenic mice were crossed with PHD2flox/flox (PHD2f/f) mice and endothelial-specific knockout of PHD2 (PHD2ECKO) mice. Transgenic PHD2f/f (TgPHD2f/f) mice and TgPHD2ECKO mice were infused with angiotensin II for 4 weeks. Arterial thickness, stiffness, and histological and immunofluorescence of pericytes and fibrosis were measured. Infusion of TgPHD2f/f mice with angiotensin II resulted in a time-dependent increase in pulse-wave velocity. Angiotensin II-induced pulse-wave velocity was further elevated in the TgPHD2ECKO mice. TgPHD2ECKO also reduced coronary flow reserve compared with TgPHD2f/f mice infused with angiotensin II. Mechanistically, knockout of endothelial PHD2 promoted aortic arginase activity and angiotensin II-induced aortic thickness together with increased transforming growth factor-β1 and ICAM-1/VCAM-1 expression in coronary arteries. TgPHD2f/f mice infused with angiotensin II for 4 weeks exhibited a significant increase in cardiac fibrosis and hypertrophy, which was further developed in the TgPHD2ECKO mice. Chondroitin sulfate proteoglycan 4 pericyte was traced by DsRed+ staining and angiotensin II infusion displayed a significant increase of DsRed+ pericytes in the heart, as well as a deficiency of endothelial PHD2, which further promoted angiotensin II-induced pericyte increase. DsRed+ pericytes were costained with fibroblast-specific protein 1 and α-smooth muscle actin for measuring pericyte-myofibroblast cell transition. The knockout of endothelial PHD2 increased the amount of DsRed+/fibroblast-specific protein 1+ and DsRed+/α-smooth muscle actin+ cells induced by angiotensin II infusion. CONCLUSIONS Knockout of endothelial PHD2 enhanced angiotensin II-induced cardiac fibrosis by mechanisms involving increasing arterial stiffness and pericyte-myofibroblast cell transitions.
Collapse
Affiliation(s)
- Bo Liu
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Heng Zeng
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Han Su
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Quinesha A. Williams
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Jessie Besanson
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Yingjie Chen
- Department of Physiology and BiophysicsUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| | - Jian‐Xiong Chen
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical Center, School of MedicineJacksonMS
| |
Collapse
|
20
|
Shimatani K, Sato H, Mizukami K, Saito A, Sasai M, Enmi JI, Watanabe K, Kamohara M, Yoshioka Y, Miyagawa S, Sawa Y. Transplantation of Human Embryonic Stem Cell-Derived Pericyte-Like Cells Transduced with Basic Fibroblast Growth Factor Promotes Angiogenic Recovery in Mice with Severe Chronic Hindlimb Ischemia. J Cardiovasc Transl Res 2024; 17:828-841. [PMID: 38376701 DOI: 10.1007/s12265-024-10496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
Critical limb ischemia (CLI) is a state of severe peripheral artery disease, with no effective treatment. Cell therapy has been investigated as a therapeutic tool for CLI, and pericytes are promising therapeutic candidates based on their angiogenic properties. We firstly generated highly proliferative and immunosuppressive pericyte-like cells from embryonic stem (ES) cells. In order to enhance the angiogenic potential, we transduced the basic fibroblast growth factor (bFGF) gene into the pericyte-like cells and found a significant enhancement of angiogenesis in a Matrigel plug assay. Furthermore, we evaluated the bFGF-expressing pericyte-like cells in the previously established chronic hindlimb ischemia model in which bone marrow-derived MSCs were not effective. As a result, bFGF-expressing pericyte-like cells significantly improved blood flow in both laser Doppler perfusion imaging (LDPI) and dynamic contrast-enhanced MRI (DCE-MRI). These findings suggest that bFGF-expressing pericyte-like cells differentiated from ES cells may be a therapeutic candidate for CLI.
Collapse
Affiliation(s)
- Kenichiro Shimatani
- Institute for Regenerative Medicine Applied Cell Therapy Research, Astellas Pharma Incorporated, 21 Miyukigaoka, Tsukuba-Shi, Ibaraki, 305-8585, Japan.
| | - Hiromu Sato
- Institute for Regenerative Medicine Applied Cell Therapy Research, Astellas Pharma Incorporated, 21 Miyukigaoka, Tsukuba-Shi, Ibaraki, 305-8585, Japan
| | - Kazuhiko Mizukami
- Institute for Regenerative Medicine Applied Cell Therapy Research, Astellas Pharma Incorporated, 21 Miyukigaoka, Tsukuba-Shi, Ibaraki, 305-8585, Japan
| | - Atsuhiro Saito
- Joint Research Chair On Design for Advanced Medical System, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masao Sasai
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jun-Ichiro Enmi
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT) and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenichi Watanabe
- Department of Cardiovascular Surgery, Hyogo Medical University Hospital, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Masazumi Kamohara
- Institute for Regenerative Medicine Applied Cell Therapy Research, Astellas Pharma Incorporated, 21 Miyukigaoka, Tsukuba-Shi, Ibaraki, 305-8585, Japan
| | - Yoshichika Yoshioka
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT) and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Future Medicine Division of Health Science, Osaka University Graduate School of Medicine, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
21
|
Hu A, Schmidt MHH, Heinig N. Microglia in retinal angiogenesis and diabetic retinopathy. Angiogenesis 2024; 27:311-331. [PMID: 38564108 PMCID: PMC11303477 DOI: 10.1007/s10456-024-09911-1] [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: 11/16/2023] [Accepted: 02/18/2024] [Indexed: 04/04/2024]
Abstract
Diabetic retinopathy has a high probability of causing visual impairment or blindness throughout the disease progression and is characterized by the growth of new blood vessels in the retina at an advanced, proliferative stage. Microglia are a resident immune population in the central nervous system, known to play a crucial role in regulating retinal angiogenesis in both physiological and pathological conditions, including diabetic retinopathy. Physiologically, they are located close to blood vessels and are essential for forming new blood vessels (neovascularization). In diabetic retinopathy, microglia become widely activated, showing a distinct polarization phenotype that leads to their accumulation around neovascular tufts. These activated microglia induce pathogenic angiogenesis through the secretion of various angiogenic factors and by regulating the status of endothelial cells. Interestingly, some subtypes of microglia simultaneously promote the regression of neovascularization tufts and normal angiogenesis in neovascularization lesions. Modulating the state of microglial activation to ameliorate neovascularization thus appears as a promising potential therapeutic approach for managing diabetic retinopathy.
Collapse
Affiliation(s)
- Aiyan Hu
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| | - Nora Heinig
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| |
Collapse
|
22
|
Moro M, Balestrero FC, Grolla AA. Pericytes: jack-of-all-trades in cancer-related inflammation. Front Pharmacol 2024; 15:1426033. [PMID: 39086395 PMCID: PMC11288921 DOI: 10.3389/fphar.2024.1426033] [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: 04/30/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Pericytes, recognized as mural cells, have long been described as components involved in blood vessel formation, playing a mere supporting role for endothelial cells (ECs). Emerging evidence strongly suggests their multifaceted roles in tissues and organs. Indeed, pericytes exhibit a remarkable ability to anticipate endothelial cell behavior and adapt their functions based on the specific cells they interact with. Pericytes can be activated by pro-inflammatory stimuli and crosstalk with immune cells, actively participating in their transmigration into blood vessels. Moreover, they can influence the immune response, often sustaining an immunosuppressive phenotype in most of the cancer types studied. In this review, we concentrate on the intricate crosstalk between pericytes and immune cells in cancer, highlighting the primary evidence regarding pericyte involvement in primary tumor mass dynamics, their contributions to tumor reprogramming for invasion and migration of malignant cells, and their role in the formation of pre-metastatic niches. Finally, we explored recent and emerging pharmacological approaches aimed at vascular normalization, including novel strategies to enhance the efficacy of immunotherapy through combined use with anti-angiogenic drugs.
Collapse
Affiliation(s)
| | | | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
23
|
Holl D, Hau WF, Julien A, Banitalebi S, Kalkitsas J, Savant S, Llorens-Bobadilla E, Herault Y, Pavlovic G, Amiry-Moghaddam M, Dias DO, Göritz C. Distinct origin and region-dependent contribution of stromal fibroblasts to fibrosis following traumatic injury in mice. Nat Neurosci 2024; 27:1285-1298. [PMID: 38849523 PMCID: PMC11239523 DOI: 10.1038/s41593-024-01678-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/10/2024] [Indexed: 06/09/2024]
Abstract
Fibrotic scar tissue formation occurs in humans and mice. The fibrotic scar impairs tissue regeneration and functional recovery. However, the origin of scar-forming fibroblasts is unclear. Here, we show that stromal fibroblasts forming the fibrotic scar derive from two populations of perivascular cells after spinal cord injury (SCI) in adult mice of both sexes. We anatomically and transcriptionally identify the two cell populations as pericytes and perivascular fibroblasts. Fibroblasts and pericytes are enriched in the white and gray matter regions of the spinal cord, respectively. Both cell populations are recruited in response to SCI and inflammation. However, their contribution to fibrotic scar tissue depends on the location of the lesion. Upon injury, pericytes and perivascular fibroblasts become activated and transcriptionally converge on the generation of stromal myofibroblasts. Our results show that pericytes and perivascular fibroblasts contribute to the fibrotic scar in a region-dependent manner.
Collapse
Affiliation(s)
- Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wing Fung Hau
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shervin Banitalebi
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jannis Kalkitsas
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Soniya Savant
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Enric Llorens-Bobadilla
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris, Illkirch-Graffenstaden, France
| | - Guillaume Pavlovic
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris, Illkirch-Graffenstaden, France
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Shatin, Hong Kong.
| |
Collapse
|
24
|
Renaud L, Wilson CL, Lafyatis R, Schnapp LM, Feghali-Bostwick CA. Transcriptomic characterization of lung pericytes in systemic sclerosis-associated pulmonary fibrosis. iScience 2024; 27:110010. [PMID: 38868196 PMCID: PMC11167435 DOI: 10.1016/j.isci.2024.110010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/09/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Systemic sclerosis (SSc) is a chronic disease characterized by fibrosis and vascular abnormalities in the skin and internal organs, including the lung. SSc-associated pulmonary fibrosis (SSc-PF) is the leading cause of death in SSc patients. Pericytes are key regulators of vascular integrity and endothelial function. The role that pericytes play in SSc-PF remains unclear. We compared the transcriptome of pericytes from SSc-PF lungs (SScL) to pericytes from normal lungs (NORML). We identified 1,179 differentially expressed genes in SScL pericytes. Pathways enriched in SScL pericytes included prostaglandin, PI3K-AKT, calcium, and vascular remodeling signaling. Decreased cyclic AMP production and altered phosphorylation of AKT in response to prostaglandin E2 in SScL pericytes demonstrate the functional consequence of changes in the prostaglandin pathway that may contribute to fibrosis. The transcriptomic signature of SSc lung pericytes suggests that they promote vascular dysfunction and contribute to the loss of protection against lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Carole L. Wilson
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Robert Lafyatis
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lynn M. Schnapp
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA
| | | |
Collapse
|
25
|
Fazio A, Neri I, Koufi FD, Marvi MV, Galvani A, Evangelisti C, McCubrey JA, Cocco L, Manzoli L, Ratti S. Signaling Role of Pericytes in Vascular Health and Tissue Homeostasis. Int J Mol Sci 2024; 25:6592. [PMID: 38928298 PMCID: PMC11203602 DOI: 10.3390/ijms25126592] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Pericytes are multipotent cells embedded within the vascular system, primarily surrounding capillaries and microvessels where they closely interact with endothelial cells. These cells are known for their intriguing properties due to their heterogeneity in tissue distribution, origin, and multifunctional capabilities. Specifically, pericytes are essential in regulating blood flow, promoting angiogenesis, and supporting tissue homeostasis and regeneration. These multifaceted roles draw on pericytes' remarkable ability to respond to biochemical cues, interact with neighboring cells, and adapt to changing environmental conditions. This review aims to summarize existing knowledge on pericytes, emphasizing their versatility and involvement in vascular integrity and tissue health. In particular, a comprehensive view of the major signaling pathways, such as PDGFβ/ PDGFRβ, TGF-β, FOXO and VEGF, along with their downstream targets, which coordinate the behavior of pericytes in preserving vascular integrity and promoting tissue regeneration, will be discussed. In this light, a deeper understanding of the complex signaling networks defining the phenotype of pericytes in healthy tissues is crucial for the development of targeted therapies in vascular and degenerative diseases.
Collapse
Affiliation(s)
- Antonietta Fazio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Irene Neri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Foteini-Dionysia Koufi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Maria Vittoria Marvi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Andrea Galvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (A.F.); (I.N.); (F.-D.K.); (M.V.M.); (A.G.); (C.E.); (L.C.); (L.M.)
| |
Collapse
|
26
|
Liao J, Gong L, Xu Q, Wang J, Yang Y, Zhang S, Dong J, Lin K, Liang Z, Sun Y, Mu Y, Chen Z, Lu Y, Zhang Q, Lin Z. Revolutionizing Neurocare: Biomimetic Nanodelivery Via Cell Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402445. [PMID: 38583077 DOI: 10.1002/adma.202402445] [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: 02/16/2024] [Revised: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Brain disorders represent a significant challenge in medical science due to the formidable blood-brain barrier (BBB), which severely limits the penetration of conventional therapeutics, hindering effective treatment strategies. This review delves into the innovative realm of biomimetic nanodelivery systems, including stem cell-derived nanoghosts, tumor cell membrane-coated nanoparticles, and erythrocyte membrane-based carriers, highlighting their potential to circumvent the BBB's restrictions. By mimicking native cell properties, these nanocarriers emerge as a promising solution for enhancing drug delivery to the brain, offering a strategic advantage in overcoming the barrier's selective permeability. The unique benefits of leveraging cell membranes from various sources is evaluated and advanced technologies for fabricating cell membrane-encapsulated nanoparticles capable of masquerading as endogenous cells are examined. This enables the targeted delivery of a broad spectrum of therapeutic agents, ranging from small molecule drugs to proteins, thereby providing an innovative approach to neurocare. Further, the review contrasts the capabilities and limitations of these biomimetic nanocarriers with traditional delivery methods, underlining their potential to enable targeted, sustained, and minimally invasive treatment modalities. This review is concluded with a perspective on the clinical translation of these biomimetic systems, underscoring their transformative impact on the therapeutic landscape for intractable brain diseases.
Collapse
Affiliation(s)
- Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lidong Gong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qingqiang Xu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Jingya Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuanyuan Yang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Shiming Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Junwei Dong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Kerui Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zichao Liang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuhan Sun
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yongxu Mu
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Ying Lu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
27
|
Suthya AR, Wong CHY, Bourne JH. Diving head-first into brain intravital microscopy. Front Immunol 2024; 15:1372996. [PMID: 38817606 PMCID: PMC11137164 DOI: 10.3389/fimmu.2024.1372996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Tissue microenvironments during physiology and pathology are highly complex, meaning dynamic cellular activities and their interactions cannot be accurately modelled ex vivo or in vitro. In particular, tissue-specific resident cells which may function and behave differently after isolation and the heterogenous vascular beds in various organs highlight the importance of observing such processes in real-time in vivo. This challenge gave rise to intravital microscopy (IVM), which was discovered over two centuries ago. From the very early techniques of low-optical resolution brightfield microscopy, limited to transparent tissues, IVM techniques have significantly evolved in recent years. Combined with improved animal surgical preparations, modern IVM technologies have achieved significantly higher speed of image acquisition and enhanced image resolution which allow for the visualisation of biological activities within a wider variety of tissue beds. These advancements have dramatically expanded our understanding in cell migration and function, especially in organs which are not easily accessible, such as the brain. In this review, we will discuss the application of rodent IVM in neurobiology in health and disease. In particular, we will outline the capability and limitations of emerging technologies, including photoacoustic, two- and three-photon imaging for brain IVM. In addition, we will discuss the use of these technologies in the context of neuroinflammation.
Collapse
|
28
|
Lin Y, Gahn J, Banerjee K, Dobreva G, Singhal M, Dubrac A, Ola R. Role of endothelial PDGFB in arterio-venous malformations pathogenesis. Angiogenesis 2024; 27:193-209. [PMID: 38070064 PMCID: PMC11021264 DOI: 10.1007/s10456-023-09900-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/05/2023] [Indexed: 04/17/2024]
Abstract
Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor β and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.
Collapse
Affiliation(s)
- Yanzhu Lin
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Gahn
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kuheli Banerjee
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montreal, QC, H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Roxana Ola
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
29
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [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: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
30
|
Ahuja S, Adjekukor C, Li Q, Kocha KM, Rosin N, Labit E, Sinha S, Narang A, Long Q, Biernaskie J, Huang P, Childs SJ. The development of brain pericytes requires expression of the transcription factor nkx3.1 in intermediate precursors. PLoS Biol 2024; 22:e3002590. [PMID: 38683849 PMCID: PMC11081496 DOI: 10.1371/journal.pbio.3002590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 05/09/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024] Open
Abstract
Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.
Collapse
Affiliation(s)
- Suchit Ahuja
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Cynthia Adjekukor
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Qing Li
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Katrinka M. Kocha
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Nicole Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Elodie Labit
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Ankita Narang
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Quan Long
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Jeff Biernaskie
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
31
|
Chakraborty MP, Das D, Mondal P, Kaul P, Bhattacharyya S, Kumar Das P, Das R. Molecular basis of VEGFR1 autoinhibition at the plasma membrane. Nat Commun 2024; 15:1346. [PMID: 38355851 PMCID: PMC10866885 DOI: 10.1038/s41467-024-45499-2] [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: 06/08/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Ligand-independent activation of VEGFRs is a hallmark of diabetes and several cancers. Like EGFR, VEGFR2 is activated spontaneously at high receptor concentrations. VEGFR1, on the other hand, remains constitutively inactive in the unligated state, making it an exception among VEGFRs. Ligand stimulation transiently phosphorylates VEGFR1 and induces weak kinase activation in endothelial cells. Recent studies, however, suggest that VEGFR1 signaling is indispensable in regulating various physiological or pathological events. The reason why VEGFR1 is regulated differently from other VEGFRs remains unknown. Here, we elucidate a mechanism of juxtamembrane inhibition that shifts the equilibrium of VEGFR1 towards the inactive state, rendering it an inefficient kinase. The juxtamembrane inhibition of VEGFR1 suppresses its basal phosphorylation even at high receptor concentrations and transiently stabilizes tyrosine phosphorylation after ligand stimulation. We conclude that a subtle imbalance in phosphatase activation or removing juxtamembrane inhibition is sufficient to induce ligand-independent activation of VEGFR1 and sustain tyrosine phosphorylation.
Collapse
Affiliation(s)
- Manas Pratim Chakraborty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Diptatanu Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Purav Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Pragya Kaul
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Soumi Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Prosad Kumar Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India
| | - Rahul Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India.
- Centre for Advanced Functional Materials, Indian Institute of Science Education and Research Kolkata, Mohanpur campus, Mohanpur, 741246, India.
| |
Collapse
|
32
|
Shen J, Chen L, Lv X, Liu N, Miao Y, Zhang Q, Xiao Z, Li M, Yang Y, Liu Z, Chen Q. Emerging Co-Assembled and Sustained Released Natural Medicinal Nanoparticles for Multitarget Therapy of Choroidal Neovascularization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2314095. [PMID: 38344832 DOI: 10.1002/adma.202314095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/29/2024] [Indexed: 02/23/2024]
Abstract
Age-related macular degeneration (AMD) disease has become a worldwide senile disease, and frequent intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) is the mainstream treatment in the clinic, which is associated with sight-threatening complications. Herein, nintedanib, an inhibitor of angiogenesis, and lutein, a potent antioxidant, can co-assemble into nanoparticles through multiple noncovalent interactions. Interestingly, the co-assembled lutein/nintedanib nanoparticles (L/N NPs) exhibit significantly improved stability and achieve long-term sustained release of two drugs for at least two months in mice. Interestingly, in rabbit eyeball with a more complete barrier system, the L/N NPs still successfully distribute in the retina and choroid for a month. In the laser-induced mouse choroidal neovascularization model, the L/N NPs after a minimally invasive subconjunctival administration can successfully inhibit angiogenesis and achieve comparable and even better therapeutic results to that of standard intravitreal injection of anti-VEGF. Therefore, the subconjunctival injection of L/N NPs with long-term sustained drug release behavior represents a promising and innovative strategy for AMD treatment. Such minimally invasive administration together with the ability to effectively inhibit angiogenesis reduce inflammation and counteract oxidative stress and holds great potential for improving patient outcomes and quality of life in those suffering from this debilitating eye condition.
Collapse
Affiliation(s)
- Jingjing Shen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Linfu Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Xinying Lv
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Nanhui Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yu Miao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qiang Zhang
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Zhisheng Xiao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Maoyi Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yang Yang
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P. R. China
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
33
|
Huang BB, Fukuyama H, Burns SA, Fawzi AA. Imaging the Retinal Vascular Mural Cells In Vivo: Elucidating the Timeline of Their Loss in Diabetic Retinopathy. Arterioscler Thromb Vasc Biol 2024; 44:465-476. [PMID: 38152885 PMCID: PMC10842708 DOI: 10.1161/atvbaha.123.320169] [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: 09/15/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Vascular mural cells (VMCs) are integral components of the retinal vasculature with critical homeostatic functions such as maintaining the inner blood-retinal barrier and vascular tone, as well as supporting the endothelial cells. Histopathologic donor eye studies have shown widespread loss of pericytes and smooth muscle cells, the 2 main VMC types, suggesting these cells are critical to the pathogenesis of diabetic retinopathy (DR). There remain, however, critical gaps in our knowledge regarding the timeline of VMC demise in human DR. METHODS In this study, we address this gap using adaptive optics scanning laser ophthalmoscopy to quantify retinal VMC density in eyes with no retinal disease (healthy), subjects with diabetes without diabetic retinopathy, and those with clinical DR and diabetic macular edema. We also used optical coherence tomography angiography to quantify capillary density of the superficial and deep capillary plexuses in these eyes. RESULTS Our results indicate significant VMC loss in retinal arterioles before the appearance of classic clinical signs of DR (diabetes without diabetic retinopathy versus healthy, 5.0±2.0 versus 6.5±2.0 smooth muscle cells per 100 µm; P<0.05), while a significant reduction in capillary VMC density (5.1±2.3 in diabetic macular edema versus 14.9±6.0 pericytes per 100 µm in diabetes without diabetic retinopathy; P=0.01) and capillary density (superficial capillary plexus vessel density, 37.6±3.8 in diabetic macular edema versus 45.5±2.4 in diabetes without diabetic retinopathy; P<0.0001) is associated with more advanced stages of clinical DR, particularly diabetic macular edema. CONCLUSIONS Our results offer a new framework for understanding the pathophysiologic course of VMC compromise in DR, which may facilitate the development and monitoring of therapeutic strategies aimed at VMC preservation and potentially the prevention of clinical DR and its associated morbidity. Imaging retinal VMCs provides an unparalleled opportunity to visualize these cells in vivo and may have wider implications in a range of diseases where these cells are disrupted.
Collapse
Affiliation(s)
- Bonnie B. Huang
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hisashi Fukuyama
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Ophthalmology, Hyogo Medical University, Hyogo, Japan
| | | | - Amani A. Fawzi
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
34
|
Kinghorn K, Gill A, Marvin A, Li R, Quigley K, Singh S, Gore MT, le Noble F, Gabhann FM, Bautch VL. A defined clathrin-mediated trafficking pathway regulates sFLT1/VEGFR1 secretion from endothelial cells. Angiogenesis 2024; 27:67-89. [PMID: 37695358 PMCID: PMC10881643 DOI: 10.1007/s10456-023-09893-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023]
Abstract
FLT1/VEGFR1 negatively regulates VEGF-A signaling and is required for proper vessel morphogenesis during vascular development and vessel homeostasis. Although a soluble isoform, sFLT1, is often mis-regulated in disease and aging, how sFLT1 is trafficked and secreted from endothelial cells is not well understood. Here we define requirements for constitutive sFLT1 trafficking and secretion in endothelial cells from the Golgi to the plasma membrane, and we show that sFLT1 secretion requires clathrin at or near the Golgi. Perturbations that affect sFLT1 trafficking blunted endothelial cell secretion and promoted intracellular mis-localization in cells and zebrafish embryos. siRNA-mediated depletion of specific trafficking components revealed requirements for RAB27A, VAMP3, and STX3 for post-Golgi vesicle trafficking and sFLT1 secretion, while STX6, ARF1, and AP1 were required at the Golgi. Live-imaging of temporally controlled sFLT1 release from the endoplasmic reticulum showed clathrin-dependent sFLT1 trafficking at the Golgi into secretory vesicles that then trafficked to the plasma membrane. Depletion of STX6 altered vessel sprouting in 3D, suggesting that endothelial cell sFLT1 secretion influences proper vessel sprouting. Thus, specific trafficking components provide a secretory path from the Golgi to the plasma membrane for sFLT1 in endothelial cells that utilizes a specialized clathrin-dependent intermediate, suggesting novel therapeutic targets.
Collapse
Affiliation(s)
- Karina Kinghorn
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Amy Gill
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Allison Marvin
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Renee Li
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Kaitlyn Quigley
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Simcha Singh
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Michaelanthony T Gore
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Picoli CDC, Birbrair A, Li Z. Pericytes as the Orchestrators of Vasculature and Adipogenesis. Genes (Basel) 2024; 15:126. [PMID: 38275607 PMCID: PMC10815550 DOI: 10.3390/genes15010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Pericytes (PCs) are located surrounding the walls of small blood vessels, particularly capillaries and microvessels. In addition to their functions in maintaining vascular integrity, participating in angiogenesis, and regulating blood flow, PCs also serve as a reservoir for multi-potent stem/progenitor cells in white, brown, beige, and bone marrow adipose tissues. Due to the complex nature of this cell population, the identification and characterization of PCs has been challenging. A comprehensive understanding of the heterogeneity of PCs may enhance their potential as therapeutic targets for metabolic syndromes or bone-related diseases. This mini-review summarizes multiple PC markers commonly employed in lineage-tracing studies, with an emphasis on their contribution to adipogenesis and functions in different adipose depots under diverse metabolic conditions.
Collapse
Affiliation(s)
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Madison, WI 53706, USA;
| | - Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA;
| |
Collapse
|
36
|
Merk L, Regel K, Eckhardt H, Evers M, El-Ayoubi A, Mittelbronn M, Krüger M, Gérardy JJ, Mack AF, Naumann U. Blocking TGF-β- and Epithelial-to-Mesenchymal Transition (EMT)-mediated activation of vessel-associated mural cells in glioblastoma impacts tumor angiogenesis. FREE NEUROPATHOLOGY 2024; 5:4. [PMID: 38455669 PMCID: PMC10919159 DOI: 10.17879/freeneuropathology-2024-5188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. GBM displays excessive and unfunctional vascularization which may, among others, be a reason for its devastating prognosis. Pericytes have been identified as the major component of the irregular vessel structure in GBM. In vitro data suggest an epithelial-to-mesenchymal transition (EMT)-like activation of glioma-associated pericytes, stimulated by GBM-secreted TGF-β, to be involved in the formation of a chaotic and dysfunctional tumor vasculature. This study investigated whether TGF-β impacts the function of vessel associated mural cells (VAMCs) in vivo via the induction of the EMT transcription factor SLUG and whether this is associated with the development of GBM-associated vascular abnormalities. Upon preventing the TGF-β-/SLUG-mediated EMT induction in VAMCs, the number of PDGFRβ and αSMA positive cells was significantly reduced, regardless of whether TGF-β secretion by GBM cells was blocked or whether SLUG was specifically knocked out in VAMCs. The reduced amount of PDGFRβ+ or αSMA+ cells observed under those conditions correlated with a lower vessel density and fewer vascular abnormalities. Our data provide evidence that the SLUG-mediated modulation of VAMC activity is induced by GBM-secreted TGF-β¬ and that activated VAMCs are key contributors in neo-angiogenic processes. We suggest that a pathologically altered activation of GA-Peris in the tumor microenvironment is responsible for the unstructured tumor vasculature. There is emerging evidence that vessel normalization alleviates tumor hypoxia, reduces tumor-associated edema and improves drug delivery. Therefore, avoiding the generation of an unstructured and non-functional tumor vasculature during tumor recurrence might be a promising treatment approach for GBM and identifies pericytes as a potential novel therapeutic target.
Collapse
Affiliation(s)
- Luisa Merk
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Katja Regel
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Hermann Eckhardt
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Marietheres Evers
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Ali El-Ayoubi
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Michel Mittelbronn
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire Nationale de Santé (LNS), Luxembourg
| | - Marcel Krüger
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Jean-Jacques Gérardy
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg
- National Center of Pathology (NCP), Laboratoire Nationale de Santé (LNS), Luxembourg
| | - Andreas F. Mack
- Institute for Clinical Anatomy and Cell Analytics, University of Tübingen, Germany
| | - Ulrike Naumann
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
- Gene and RNA Therapy Center (GRTC), Faculty of Medicine University Tübingen, Germany
| |
Collapse
|
37
|
van Splunder H, Villacampa P, Martínez-Romero A, Graupera M. Pericytes in the disease spotlight. Trends Cell Biol 2024; 34:58-71. [PMID: 37474376 PMCID: PMC10777571 DOI: 10.1016/j.tcb.2023.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 07/22/2023]
Abstract
Pericytes are known as the mural cells in small-caliber vessels that interact closely with the endothelium. Pericytes play a key role in vasculature formation and homeostasis, and when dysfunctional contribute to vasculature-related diseases such as diabetic retinopathy and neurodegenerative conditions. In addition, significant extravascular roles of pathological pericytes are being discovered with relevant implications for cancer and fibrosis. Pericyte research is challenged by the lack of consistent molecular markers and clear discrimination criteria versus other (mural) cells. However, advances in single-cell approaches are uncovering and clarifying mural cell identities, biological functions, and ontogeny across organs. We discuss the latest developments in pericyte pathobiology to inform future research directions and potential outcomes.
Collapse
Affiliation(s)
- Hielke van Splunder
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Pilar Villacampa
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Carrer de la Feixa Llarga s/n, 08907 l'Hospitalet de Llobregat, Barcelona, Spain
| | - Anabel Martínez-Romero
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain; Institución Catalana de Investigación y Estudios Avanzados (ICREA), Passeig de Lluís Companys 23, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Avenida de Monforte de Lemos 5, 28029 Madrid, Spain.
| |
Collapse
|
38
|
You TY, Dong Q, Cui M. Emerging Links between Cerebral Blood Flow Regulation and Cognitive Decline: A Role for Brain Microvascular Pericytes. Aging Dis 2023:AD.2022.1204. [PMID: 37163446 PMCID: PMC10389833 DOI: 10.14336/ad.2022.1204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/04/2022] [Indexed: 05/12/2023] Open
Abstract
Cognitive impairment associated with vascular etiology has been of considerable interest in the development of dementia. Recent studies have started to uncover cerebral blood flow deficits in initiating cognitive deterioration. Brain microvascular pericytes, the only type of contractile cells in capillaries, are involved in the precise modulation of vascular hemodynamics due to their ability to regulate resistance in the capillaries. They exhibit potential in maintaining the capillary network geometry and basal vascular tone. In addition, pericytes can facilitate better blood flow supply in response to neurovascular coupling. Their dysfunction is thought to disturb cerebral blood flow causing metabolic imbalances or structural injuries, leading to consequent cognitive decline. In this review, we summarize the characteristics of microvascular pericytes in brain blood flow regulation and outline the framework of a two-hit hypothesis in cognitive decline, where we emphasize how pericytes serve as targets of cerebral blood flow dysregulation that occurs with neurological challenges, ranging from genetic factors, aging, and pathological proteins to ischemic stress.
Collapse
Affiliation(s)
- Tong-Yao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Boss AL, Chamley LW, Brooks AES, James JL. Human placental vascular and perivascular cell heterogeneity differs between first trimester and term, and in pregnancies affected by foetal growth restriction. Mol Hum Reprod 2023; 29:gaad041. [PMID: 38059603 PMCID: PMC10746841 DOI: 10.1093/molehr/gaad041] [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: 05/25/2023] [Revised: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Growth-restricted placentae have a reduced vascular network, impairing exchange of nutrients and oxygen. However, little is known about the differentiation events and cell types that underpin normal/abnormal placental vascular formation and function. Here, we used 23-colour flow cytometry to characterize placental vascular/perivascular populations between first trimester and term, and in foetal growth restriction (FGR). First-trimester endothelial cells had an immature phenotype (CD144+/lowCD36-CD146low), while term endothelial cells expressed mature endothelial markers (CD36+CD146+). At term, a distinct population of CD31low endothelial cells co-expressed mesenchymal markers (CD90, CD26), indicating a capacity for endothelial to mesenchymal transition (EndMT). In FGR, compared with normal pregnancies, endothelial cells constituted 3-fold fewer villous core cells (P < 0.05), contributing to an increased perivascular: endothelial cell ratio (2.6-fold, P < 0.05). This suggests that abnormal EndMT may play a role in FGR. First-trimester endothelial cells underwent EndMT in culture, losing endothelial (CD31, CD34, CD144) and gaining mesenchymal (CD90, CD26) marker expression. Together this highlights how differences in villous core cell heterogeneity and phenotype may contribute to FGR pathophysiology across gestation.
Collapse
Affiliation(s)
- Anna L Boss
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
40
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
41
|
Zoungrana LI, Didik S, Wang H, Slotabec L, Li J. Activated protein C in epilepsy pathophysiology. Front Neurosci 2023; 17:1251017. [PMID: 37901428 PMCID: PMC10603301 DOI: 10.3389/fnins.2023.1251017] [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: 06/30/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Epilepsy is one of the most common neurologic disorders that is characterized by recurrent seizures, and depending on the type of seizure, it could lead to a severe outcome. Epilepsy's mechanism of development is not fully understood yet, but some of the common features of the disease are blood-brain barrier disruption, microglia activation, and neuroinflammation. Those are also targets of activated protein C (APC). In fact, by downregulating thrombin, known as a pro-inflammatory, APC acts as an anti-inflammatory. APC is also an anti-apoptotic protein, instance by blocking p53-mediated apoptosis. APC's neuroprotective effect could prevent blood-brain barrier dysfunction by acting on endothelial cells. Furthermore, through the downregulation of proapoptotic, and proinflammatory genes, APC's neuroprotection could reduce the effect or prevent epilepsy pathogenesis. APC's activity acts on blood-brain barrier disruption, inflammation, and apoptosis and causes neurogenesis, all hallmarks that could potentially treat or prevent epilepsy. Here we review both Activated Protein C and epilepsy mechanism, function, and the possible association between them.
Collapse
Affiliation(s)
- Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Steven Didik
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Hao Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lily Slotabec
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ji Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
42
|
Bora K, Kushwah N, Maurya M, Pavlovich MC, Wang Z, Chen J. Assessment of Inner Blood-Retinal Barrier: Animal Models and Methods. Cells 2023; 12:2443. [PMID: 37887287 PMCID: PMC10605292 DOI: 10.3390/cells12202443] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Proper functioning of the neural retina relies on the unique retinal environment regulated by the blood-retinal barrier (BRB), which restricts the passage of solutes, fluids, and toxic substances. BRB impairment occurs in many retinal vascular diseases and the breakdown of BRB significantly contributes to disease pathology. Understanding the different molecular constituents and signaling pathways involved in BRB development and maintenance is therefore crucial in developing treatment modalities. This review summarizes the major molecular signaling pathways involved in inner BRB (iBRB) formation and maintenance, and representative animal models of eye diseases with retinal vascular leakage. Studies on Wnt/β-catenin signaling are highlighted, which is critical for retinal and brain vascular angiogenesis and barriergenesis. Moreover, multiple in vivo and in vitro methods for the detection and analysis of vascular leakage are described, along with their advantages and limitations. These pre-clinical animal models and methods for assessing iBRB provide valuable experimental tools in delineating the molecular mechanisms of retinal vascular diseases and evaluating therapeutic drugs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
43
|
Pauzuolyte V, Patel A, Wawrzynski JR, Ingham NJ, Leong YC, Karda R, Bitner‐Glindzicz M, Berger W, Waddington SN, Steel KP, Sowden JC. Systemic gene therapy rescues retinal dysfunction and hearing loss in a model of Norrie disease. EMBO Mol Med 2023; 15:e17393. [PMID: 37642150 PMCID: PMC10565640 DOI: 10.15252/emmm.202317393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Deafness affects 5% of the world's population, yet there is a lack of treatments to prevent hearing loss due to genetic causes. Norrie disease is a recessive X-linked disorder, caused by NDP gene mutation. It manifests as blindness at birth and progressive sensorineural hearing loss, leading to debilitating dual sensory deprivation. To develop a gene therapy, we used a Norrie disease mouse model (Ndptm1Wbrg ), which recapitulates abnormal retinal vascularisation and progressive hearing loss. We delivered human NDP cDNA by intravenous injection of adeno-associated viral vector (AAV)9 at neonatal, juvenile and young adult pathological stages and investigated its therapeutic effects on the retina and cochlea. Neonatal treatment prevented the death of the sensory cochlear hair cells and rescued cochlear disease biomarkers as demonstrated by RNAseq and physiological measurements of auditory function. Retinal vascularisation and electroretinograms were restored to normal by neonatal treatment. Delivery of NDP gene therapy after the onset of the degenerative inner ear disease also ameliorated the cochlear pathology, supporting the feasibility of a clinical treatment for progressive hearing loss in people with Norrie disease.
Collapse
Affiliation(s)
- Valda Pauzuolyte
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Aara Patel
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - James R Wawrzynski
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Neil J Ingham
- Wolfson Centre for Age‐Related Diseases, King's College LondonLondonUK
| | - Yeh Chwan Leong
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Rajvinder Karda
- EGA Institute for Woman's Health, University College LondonLondonUK
| | - Maria Bitner‐Glindzicz
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Wolfgang Berger
- Institute of Medical Molecular Genetics, University of ZürichZürichSwitzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of ZürichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH Zurich, University of ZürichZürichSwitzerland
| | - Simon N Waddington
- EGA Institute for Woman's Health, University College LondonLondonUK
- MRC Antiviral Gene Therapy Research Unit, Faculty of Health SciencesUniversity of the WitswatersrandJohannesburgSouth Africa
| | - Karen P Steel
- Wolfson Centre for Age‐Related Diseases, King's College LondonLondonUK
| | - Jane C Sowden
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| |
Collapse
|
44
|
Shi X. Research advances in cochlear pericytes and hearing loss. Hear Res 2023; 438:108877. [PMID: 37651921 PMCID: PMC10538405 DOI: 10.1016/j.heares.2023.108877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/03/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Pericytes are specialized mural cells surrounding endothelial cells in microvascular beds. They play a role in vascular development, blood flow regulation, maintenance of blood-tissue barrier integrity, and control of angiogenesis, tissue fibrosis, and wound healing. In recent decades, understanding of the critical role played by pericytes in retina, brain, lung, and kidney has seen significant progress. The cochlea contains a large population of pericytes. However, the role of cochlear pericytes in auditory pathophysiology is, by contrast, largely unknown. The present review discusses recent progress in identifying cochlear pericytes, mapping their distribution, and defining their role in regulating blood flow, controlling the blood-labyrinth barrier (BLB) and angiogenesis, and involvement in different types of hearing loss.
Collapse
Affiliation(s)
- Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center (NRC04), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| |
Collapse
|
45
|
Guo M, Wikenheiser-Brokamp KA, Kitzmiller JA, Jiang C, Wang G, Wang A, Preissl S, Hou X, Buchanan J, Karolak JA, Miao Y, Frank DB, Zacharias WJ, Sun X, Xu Y, Gu M, Stankiewicz P, Kalinichenko VV, Wambach JA, Whitsett JA. Single Cell Multiomics Identifies Cells and Genetic Networks Underlying Alveolar Capillary Dysplasia. Am J Respir Crit Care Med 2023; 208:709-725. [PMID: 37463497 PMCID: PMC10515568 DOI: 10.1164/rccm.202210-2015oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/18/2023] [Indexed: 07/20/2023] Open
Abstract
Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.
Collapse
Affiliation(s)
- Minzhe Guo
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Department of Pediatrics and
| | - Kathryn A. Wikenheiser-Brokamp
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Division of Pathology and Laboratory Medicine
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Joseph A. Kitzmiller
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
| | - Cheng Jiang
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
| | - Guolun Wang
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Center for Lung Regenerative Medicine
| | - Allen Wang
- Center for Epigenomics & Department of Cellular & Molecular Medicine
| | - Sebastian Preissl
- Center for Epigenomics & Department of Cellular & Molecular Medicine
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xiaomeng Hou
- Center for Epigenomics & Department of Cellular & Molecular Medicine
| | - Justin Buchanan
- Center for Epigenomics & Department of Cellular & Molecular Medicine
| | - Justyna A. Karolak
- Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Yifei Miao
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Division of Developmental Biology, and
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics and
| | - David B. Frank
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - William J. Zacharias
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Department of Pediatrics and
| | - Xin Sun
- Department of Pediatrics, and
- Department of Biological Sciences, University of California, San Diego, La Jolla, California
| | - Yan Xu
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Division of Biomedical Informatics
- Department of Pediatrics and
| | - Mingxia Gu
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Division of Developmental Biology, and
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics and
| | - Pawel Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas; and
| | - Vladimir V. Kalinichenko
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Center for Lung Regenerative Medicine
- Department of Pediatrics and
| | - Jennifer A. Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri
| | - Jeffrey A. Whitsett
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology
- Department of Pediatrics and
| |
Collapse
|
46
|
Fu J, Liang H, Yuan P, Wei Z, Zhong P. Brain pericyte biology: from physiopathological mechanisms to potential therapeutic applications in ischemic stroke. Front Cell Neurosci 2023; 17:1267785. [PMID: 37780206 PMCID: PMC10536258 DOI: 10.3389/fncel.2023.1267785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Pericytes play an indispensable role in various organs and biological processes, such as promoting angiogenesis, regulating microvascular blood flow, and participating in immune responses. Therefore, in this review, we will first introduce the discovery and development of pericytes, identification methods and functional characteristics, then focus on brain pericytes, on the one hand, to summarize the functions of brain pericytes under physiological conditions, mainly discussing from the aspects of stem cell characteristics, contractile characteristics and paracrine characteristics; on the other hand, to summarize the role of brain pericytes under pathological conditions, mainly taking ischemic stroke as an example. Finally, we will discuss and analyze the application and development of pericytes as therapeutic targets, providing the research basis and direction for future microvascular diseases, especially ischemic stroke treatment.
Collapse
Affiliation(s)
- Jiaqi Fu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou, Jiangsu, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenyu Wei
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Ping Zhong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
47
|
Tatsukawa T, Kano K, Nakajima KI, Yazawa T, Eguchi R, Kabara M, Horiuchi K, Hayasaka T, Matsuo R, Hasebe N, Azuma N, Kawabe JI. NG2-positive pericytes regulate homeostatic maintenance of slow-type skeletal muscle with rapid myonuclear turnover. Stem Cell Res Ther 2023; 14:205. [PMID: 37592340 PMCID: PMC10433572 DOI: 10.1186/s13287-023-03433-1] [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: 01/24/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Skeletal muscle comprises almost 40% of the human body and is essential for movement, structural support and metabolic homeostasis. Size of multinuclear skeletal muscle is stably maintained under steady conditions with the sporadic fusion of newly produced myocytes to compensate for the muscular turnover caused by daily wear and tear. It is becoming clear that microvascular pericytes (PCs) exhibit myogenic activity. However, whether PCs act as myogenic stem cells for the homeostatic maintenance of skeletal muscles during adulthood remains uncertain. METHODS We utilized PC-fused myofibers using PC-specific lineage tracing mouse (NG2-CreERT/Rosa-tdTomato) to observe whether muscle resident PCs have myogenic potential during daily life. Genetic PC deletion mouse model (NG2-CreERT/DTA) was used to test whether PC differentiates to myofibers for maintenance of muscle structure and function under homeostatic condition. RESULTS Under steady breeding conditions, tdTomato-expressing PCs were infused into myofibers, and subsequently, PC-derived nuclei were incorporated into myofibers. Especially in type-I slow-type myofibers such as the soleus, tdTomato+ myofibers were already observed 3 days after PC labeling; their ratio reached a peak (approximately 80%) within 1 month and was maintained for more than 1 year. Consistently, the NG2+ PC-specific deletion induced muscular atrophy in a slow-type myofiber-specific manner under steady breeding conditions. The number of myonucleus per volume of each myofiber was constant during observation period. CONCLUSIONS These findings demonstrate that the turnover of myonuclei in slow-type myofibers is relatively fast, with PCs acting as myogenic stem cells-the suppliers of new myonuclei under steady conditions-and play a vital role in the homeostatic maintenance of slow-type muscles.
Collapse
Affiliation(s)
- Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Dermatology, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Naoyuki Hasebe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
48
|
Abstract
The vasculature consists of vessels of different sizes that are arranged in a hierarchical pattern. Two cell populations work in concert to establish this pattern during embryonic development and adopt it to changes in blood flow demand later in life: endothelial cells that line the inner surface of blood vessels, and adjacent vascular mural cells, including smooth muscle cells and pericytes. Despite recent progress in elucidating the signalling pathways controlling their crosstalk, much debate remains with regard to how mural cells influence endothelial cell biology and thereby contribute to the regulation of blood vessel formation and diameters. In this Review, I discuss mural cell functions and their interactions with endothelial cells, focusing on how these interactions ensure optimal blood flow patterns. Subsequently, I introduce the signalling pathways controlling mural cell development followed by an overview of mural cell ontogeny with an emphasis on the distinguishing features of mural cells located on different types of blood vessels. Ultimately, I explore therapeutic strategies involving mural cells to alleviate tissue ischemia and improve vascular efficiency in a variety of diseases.
Collapse
Affiliation(s)
- Arndt F. Siekmann
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 1114 Biomedical Research Building, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Günter A, Sothilingam V, Orlich MM, Nordheim A, Seeliger MW, Mühlfriedel R. Mural Serum Response Factor (SRF) Deficiency Provides Insights into Retinal Vascular Functionality and Development. Int J Mol Sci 2023; 24:12597. [PMID: 37628776 PMCID: PMC10454173 DOI: 10.3390/ijms241612597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Serum response factor (SRF) controls the expression of muscle contraction and motility genes in mural cells (MCs) of the vasculature. In the retina, MC-SRF is important for correct angiogenesis during development and the continuing maintenance of the vascular tone. The purpose of this study was to provide further insights into the effects of MC SRF deficiency on the vasculature and function of the mature retina in SrfiMCKO mice that carry a MC-specific deletion of Srf. Retinal morphology and vascular integrity were analyzed in vivo via scanning laser ophthalmoscopy (SLO), angiography, and optical coherence tomography (OCT). Retinal function was evaluated with full-field electroretinography (ERG). We found that retinal blood vessels of these mutants exhibited different degrees of morphological and functional alterations. With increasing severity, we found vascular bulging, the formation of arteriovenous (AV) anastomoses, and ultimately, a retinal detachment (RD). The associated irregular retinal blood pressure and flow distribution eventually induced hypoxia, indicated by a negative ERG waveform shape. Further, the high frequency of interocular differences in the phenotype of individual SrfiMCKO mice points to a secondary nature of these developments far downstream of the genetic defect and rather dependent on the local retinal context.
Collapse
Affiliation(s)
- Alexander Günter
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (V.S.); (M.W.S.)
| | - Vithiyanjali Sothilingam
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (V.S.); (M.W.S.)
| | - Michael M. Orlich
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden;
| | - Alfred Nordheim
- Department of Molecular Biology, Interfaculty Institute of Cell Biology, University of Tübingen, 72076 Tübingen, Germany;
| | - Mathias W. Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (V.S.); (M.W.S.)
| | - Regine Mühlfriedel
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (V.S.); (M.W.S.)
| |
Collapse
|
50
|
Wu Y, Fu J, Huang Y, Duan R, Zhang W, Wang C, Wang S, Hu X, Zhao H, Wang L, Liu J, Gao G, Yuan P. Biology and function of pericytes in the vascular microcirculation. Animal Model Exp Med 2023; 6:337-345. [PMID: 37317664 PMCID: PMC10486323 DOI: 10.1002/ame2.12334] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/16/2023] [Indexed: 06/16/2023] Open
Abstract
Pericytes are the main cellular components of tiny arteries and capillaries. Studies have found that pericytes can undergo morphological contraction or relaxation under stimulation by cytokines, thus affecting the contraction and relaxation of microvessels and playing an essential role in regulating vascular microcirculation. Moreover, due to the characteristics of stem cells, pericytes can differentiate into a variety of inflammatory cell phenotypes, which then affect the immune function. Additionally, pericytes can also participate in angiogenesis and wound healing by interacting with endothelial cells in vascular microcirculation disorders. Here we review the origin, biological phenotype and function of pericytes, and discuss the potential mechanisms of pericytes in vascular microcirculation disorders, especially in pulmonary hypertension, so as to provide a sound basis and direction for the prevention and treatment of vascular microcirculation diseases.
Collapse
Affiliation(s)
- Yue Wu
- Ningbo University School of MedicineNingboChina
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jiaqi Fu
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
- Institute of Health Science and EngineeringUniversity of Shanghai Science and TechnologyShanghaiChina
| | - Yuxia Huang
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Ruowang Duan
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Wentian Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Caihong Wang
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
- Institute of Bismuth ScienceUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Shang Wang
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiaoyi Hu
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Hui Zhao
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
- Institute of Bismuth ScienceUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Lan Wang
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jinming Liu
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Guosheng Gao
- Ningbo Huamei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Ping Yuan
- Department of Cardio‐Pulmonary Circulation, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|