1
|
Couto M, Vasconcelos DP, Pereira CL, Neto E, Sarmento B, Lamghari M. Neuro-Immunomodulatory Potential of Nanoenabled 4D Bioprinted Microtissue for Cartilage Tissue Engineering. Adv Healthc Mater 2024:e2400496. [PMID: 38850170 DOI: 10.1002/adhm.202400496] [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: 02/07/2024] [Revised: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Cartilage defects trigger post-traumatic inflammation, leading to a catabolic metabolism in chondrocytes and exacerbating cartilage degradation. Current treatments aim to relieve pain but fail to target the inflammatory process underlying osteoarthritis (OA) progression. Here, a human cartilage microtissue (HCM) nanoenabled with ibuprofen-loaded poly(lactic-co-glycolic acid) nanoparticles (ibu-PLGA NPs) is 4D-bioprinted to locally mitigate inflammation and impair nerve sprouting. Under an in vitro inflamed environment, the nanoenabled HCM exhibits chondroprotective potential by decreasing the interleukin (IL)1β and IL6 release, while sustaining extracellular matrix (ECM) production. In vivo, assessments utilizing the air pouch mouse model affirm the nanoenabled HCM non-immunogenicity. Nanoenabled HCM-derived secretomes do not elicit a systemic immune response and decrease locally the recruitment of mature dendritic cells and the secretion of multiple inflammatory mediators and matrix metalloproteinases when compared to inflamed HCM condition. Notably, the nanoenabled HCM secretome has no impact on the innervation profile of the skin above the pouch cavity, suggesting a potential to impede nerve growth. Overall, HCM nanoenabled with ibu-PLGA NPs emerges as a potent strategy to mitigate inflammation and protect ECM without triggering nerve growth, introducing an innovative and promising approach in the cartilage tissue engineering field.
Collapse
Affiliation(s)
- Marina Couto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto - ICBAS, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Daniela Pereira Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
| | - Estrela Neto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, Porto, 4200-072, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- Instituto Universitário de Ciências da Saúde - IUCS-CESPU, Rua Central de Gandra, 1317, Gandra, 4585-116, Portugal
| | - Meriem Lamghari
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
- INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-125, Portugal
| |
Collapse
|
2
|
Zhang X, Duan X, Liu X. The role of kinases in peripheral nerve regeneration: mechanisms and implications. Front Neurol 2024; 15:1340845. [PMID: 38689881 PMCID: PMC11058862 DOI: 10.3389/fneur.2024.1340845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Peripheral nerve injury disease is a prevalent traumatic condition in current medical practice. Despite the present treatment approaches, encompassing surgical sutures, autologous nerve or allograft nerve transplantation, tissue engineering techniques, and others, an effective clinical treatment method still needs to be discovered. Exploring novel treatment methods to improve peripheral nerve regeneration requires more effort in investigating the cellular and molecular mechanisms involved. Many factors are associated with the regeneration of injured peripheral nerves, including the cross-sectional area of the injured nerve, the length of the nerve gap defect, and various cellular and molecular factors such as Schwann cells, inflammation factors, kinases, and growth factors. As crucial mediators of cellular communication, kinases exert regulatory control over numerous signaling cascades, thereby participating in various vital biological processes, including peripheral nerve regeneration after nerve injury. In this review, we examined diverse kinase classifications, distinct nerve injury types, and the intricate mechanisms involved in peripheral nerve regeneration. Then we stressed the significance of kinases in regulating autophagy, inflammatory response, apoptosis, cell cycle, oxidative processes, and other aspects in establishing conductive microenvironments for nerve tissue regeneration. Finally, we briefly discussed the functional roles of kinases in different types of cells involved in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xu Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, School of Life Science, Nantong Laboratory of Development and Diseases, Medical College, Clinical Medical Research Center, Affiliated Wuxi Clinical College of Nantong University, Nantong University, Nantong, China
- Clinical Medical Research Center, Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, Wuxi, China
| | - Xuchu Duan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, School of Life Science, Nantong Laboratory of Development and Diseases, Medical College, Clinical Medical Research Center, Affiliated Wuxi Clinical College of Nantong University, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, School of Life Science, Nantong Laboratory of Development and Diseases, Medical College, Clinical Medical Research Center, Affiliated Wuxi Clinical College of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
3
|
Agyekum JA, Yeboah K. Peripheral sensory neuropathy is associated with circulating angiopoietins in type 2 diabetes patients in Ghana. J Clin Transl Endocrinol 2023; 34:100327. [PMID: 37822668 PMCID: PMC10563058 DOI: 10.1016/j.jcte.2023.100327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/09/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
Objective Peripheral sensory neuropathy (PSN) is a common complication of type 2 diabetes (T2DM) that can lead to frequent ulcerations, lower extremities, and reduced quality of life. Imbalance in the circulating levels of angiogenic growth factors, notably, angiopoietin (Ang)-1, Ang-2 and vascular endothelial growth factor (VEGF) may be among the underlying mechanisms of PSN in T2DM patients. We studied the association between PSN and angiogenic growth factors, Ang-1, Ang-2 and VEGF in T2DM patients in Ghana. Methods In a case-control study design, PSN was evaluated in 160 patients with T2DM and 108 nondiabetic controls using vibration perception threshold (VPT) and diabetic neurological examination (DNE). The definition of PSN was abnormal VPT (≥25 mV) or the presence of neuropathic symptoms on examination (DNE score > 3). In addition, fasting venous blood samples were collected to measure circulating levels of Ang-1, Ang-2 and VEGF. Results Compared to non-diabetic controls, patients with T2DM had a higher prevalence of PSN using abnormal VPT (20.6 % vs 2.8 %, p < 0.001) or neuropathic symptoms (35.6 % vs 3.7 %, p < 0.001). Compared to nondiabetic controls, patients with T2DM had increased levels of Ang-2 [597 (274 - 1005) vs 838 (473 - 1241) ng/ml, p = 0.018] and VEGF [48.4 (17.4 - 110.1) vs 72.2 (28 - 201.8), p = 0.025] and decreased Ang-1 levels [41.1 (30 - 57.3) vs 36.1 (24.7 - 42.1) ng/ml, p = 0.01]. In regression analyses, an increase in Ang-1 levels was associated with decreased odds, while an increase in Ang-2 levels was associated with increased odds, of abnormal VPT and neuropathic symptoms in T2DM patients. Conclusion In our study population, PSN was associated with reduced plasma levels of Ang-1 and increased plasma levels of Ang-2 in patients with T2DM. Therefore, an imbalance of angiopoietins may be associated with PSN in T2DM.
Collapse
Affiliation(s)
- Jennifer A. Agyekum
- Department of Physiology, University of Ghana Medical School, Accra, Ghana
- Medical Laboratory Unit, Mamprobi Hospital, Ghana Health Services, Accra, Ghana
| | - Kwame Yeboah
- Department of Physiology, University of Ghana Medical School, Accra, Ghana
| |
Collapse
|
4
|
Dieter C, Lemos NE, de Faria Corrêa NR, Assmann TS, Pellenz FM, Canani LH, de Almeida Brondani L, Bauer AC, Crispim D. Polymorphisms in TIE2 and ANGPT-1 genes are associated with protection against diabetic retinopathy in a Brazilian population. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:e000624. [PMID: 37249455 PMCID: PMC10665047 DOI: 10.20945/2359-3997000000624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 12/06/2022] [Indexed: 05/31/2023]
Abstract
Objective The objective of this study was to investigate the association between SNPs in the TIE2 and ANGPT-1 genes and diabetic retinopathy (DR). Subjects and methods This study comprised 603 patients with type 2 diabetes mellitus (T2DM) and DR (cases) and 388 patients with T2DM for more than 10 years and without DR (controls). The TIE2 rs639225 (A/G) and rs638203 (A/G) SNPs and the ANGPT-1 rs4324901 (G/T) and rs2507800 (T/A) SNPs were genotyped by real-time PCR using TaqMan MGB probes. Results The G/G genotype of the rs639225/TIE2, the G/G genotype of the rs638203/ TIE2 and the T allele of the rs4324901/ANGPT-1 SNPs were associated with protection against DR after adjustment for age, glycated hemoglobin, gender, and presence of hypertension (P = 0.042, P = 0.003, and P = 0.028, respectively). No association was found between the rs2507800/ANGPT-1 SNP and DR. Conclusion We demonstrated, for the first time, the association of TIE2 rs638203 and rsrs939225 SNPs and ANGPT-1 rs4324901 SNP with protection against DR in a Brazilian population.
Collapse
Affiliation(s)
- Cristine Dieter
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | - Natália Emerim Lemos
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | | | - Taís Silveira Assmann
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | - Felipe Mateus Pellenz
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | - Luís Henrique Canani
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | | | - Andrea Carla Bauer
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Hospital de Clínicas de Porto Alegre, Divisão de Nefrologia, Porto Alegre, RS, Brasil
| | - Daisy Crispim
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil,
| |
Collapse
|
5
|
Kim D, Kim S, Sung A, Patel N, Wong N, Conboy MJ, Conboy IM. Autologous treatment for ALS with implication for broad neuroprotection. Transl Neurodegener 2022; 11:16. [PMID: 35272709 PMCID: PMC8915496 DOI: 10.1186/s40035-022-00290-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/18/2022] [Indexed: 01/20/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is characterized by a progressive loss of motor neurons (MNs), leading to paralysis, respiratory failure and death within 2–5 years of diagnosis. The exact mechanisms of sporadic ALS, which comprises 90% of all cases, remain unknown. In familial ALS, mutations in superoxide dismutase (SOD1) cause 10% of cases. Methods ALS patient-derived human-induced pluripotent stem cells (ALS hiPSCs, harboring the SOD1AV4 mutation), were differentiated to MNs (ALS-MNs). The neuroprotective effects of conditioned medium (CM) of hESCs (H9), wt hiPSCs (WTC-11) and the ALS iPSCs, on MN apoptosis and viability, formation and maintenance of neurites, mitochondrial activity and expression of inflammatory genes, were examined. For in vivo studies, 200 μl of CM from the ALS iPSCs (CS07 and CS053) was injected subcutaneously into the ALS model mice (transgenic for the human SOD1G93A mutation). Animal agility and strength, muscle innervation and mass, neurological score, onset of paralysis and lifespan of the ALS mice were assayed. After observing significant disease-modifying effects, the CM was characterized biochemically by fractionation, comparative proteomics, and epigenetic screens for the dependence on pluripotency. CM of fibroblasts that were differentiated from the wt hiPSCs lacked any neuroprotective activity and was used as a negative control throughout the studies. Results The secretome of PSCs including the ALS patient iPSCs was neuroprotective in the H2O2 model. In the model with pathogenic SOD1 mutation, ALS iPSC-CM attenuated all examined hallmarks of ALS pathology, rescued human ALS-MNs from denervation and death, restored mitochondrial health, and reduced the expression of inflammatory genes. The ALS iPSC-CM also improved neuro-muscular health and function, and delayed paralysis and morbidity in ALS mice. Compared side by side, cyclosporine (CsA), a mitochondrial membrane blocker that prevents the leakage of mitochondrial DNA, failed to avert the death of ALS-MNs, although CsA and ALS iPSC-CM equally stabilized MN mitochondria and attenuated inflammatory genes. Biochemical characterization, comparative proteomics, and epigenetic screen all suggested that it was the interactome of several key proteins from different fractions of PSC-CM that delivered the multifaceted neuroprotection. Conclusions This work introduces and mechanistically characterizes a new biologic for treating ALS and other complex neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00290-5.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Subin Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Ashley Sung
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Neetika Patel
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Nathan Wong
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
6
|
Luck R, Karakatsani A, Shah B, Schermann G, Adler H, Kupke J, Tisch N, Jeong HW, Back MK, Hetsch F, D'Errico A, De Palma M, Wiedtke E, Grimm D, Acker-Palmer A, von Engelhardt J, Adams RH, Augustin HG, Ruiz de Almodóvar C. The angiopoietin-Tie2 pathway regulates Purkinje cell dendritic morphogenesis in a cell-autonomous manner. Cell Rep 2021; 36:109522. [PMID: 34407407 PMCID: PMC9110807 DOI: 10.1016/j.celrep.2021.109522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/06/2021] [Accepted: 07/22/2021] [Indexed: 01/01/2023] Open
Abstract
Neuro-vascular communication is essential to synchronize central nervous system development. Here, we identify angiopoietin/Tie2 as a neuro-vascular signaling axis involved in regulating dendritic morphogenesis of Purkinje cells (PCs). We show that in the developing cerebellum Tie2 expression is not restricted to blood vessels, but it is also present in PCs. Its ligands angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are expressed in neural cells and endothelial cells (ECs), respectively. PC-specific deletion of Tie2 results in reduced dendritic arborization, which is recapitulated in neural-specific Ang1-knockout and Ang2 full-knockout mice. Mechanistically, RNA sequencing reveals that Tie2-deficient PCs present alterations in gene expression of multiple genes involved in cytoskeleton organization, dendritic formation, growth, and branching. Functionally, mice with deletion of Tie2 in PCs present alterations in PC network functionality. Altogether, our data propose Ang/Tie2 signaling as a mediator of intercellular communication between neural cells, ECs, and PCs, required for proper PC dendritic morphogenesis and function.
Collapse
Affiliation(s)
- Robert Luck
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Andromachi Karakatsani
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Bhavin Shah
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Geza Schermann
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Heike Adler
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), University of Heidelberg, 69120 Heidelberg, Germany
| | - Nathalie Tisch
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and University of Münster, Faculty of Medicine, 48149 Münster, Germany
| | - Michaela Kerstin Back
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Florian Hetsch
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Anna D'Errico
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60323 Frankfurt, Germany
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ellen Wiedtke
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60323 Frankfurt, Germany
| | - Jakob von Engelhardt
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and University of Münster, Faculty of Medicine, 48149 Münster, Germany
| | - Hellmut G Augustin
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Carmen Ruiz de Almodóvar
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| |
Collapse
|
7
|
Sala-Jarque J, Mesquida-Veny F, Badiola-Mateos M, Samitier J, Hervera A, del Río JA. Neuromuscular Activity Induces Paracrine Signaling and Triggers Axonal Regrowth after Injury in Microfluidic Lab-On-Chip Devices. Cells 2020; 9:cells9020302. [PMID: 32012727 PMCID: PMC7072511 DOI: 10.3390/cells9020302] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Peripheral nerve injuries, including motor neuron axonal injury, often lead to functional impairments. Current therapies are mostly limited to surgical intervention after lesion, yet these interventions have limited success in restoring functionality. Current activity-based therapies after axonal injuries are based on trial-error approaches in which the details of the underlying cellular and molecular processes are largely unknown. Here we show the effects of the modulation of both neuronal and muscular activity with optogenetic approaches to assess the regenerative capacity of cultured motor neuron (MN) after lesion in a compartmentalized microfluidic-assisted axotomy device. With increased neuronal activity, we observed an increase in the ratio of regrowing axons after injury in our peripheral-injury model. Moreover, increasing muscular activity induces the liberation of leukemia inhibitory factor and glial cell line-derived neurotrophic factor in a paracrine fashion that in turn triggers axonal regrowth of lesioned MN in our 3D hydrogel cultures. The relevance of our findings as well as the novel approaches used in this study could be useful not only after axotomy events but also in diseases affecting MN survival.
Collapse
Affiliation(s)
- Julia Sala-Jarque
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Francina Mesquida-Veny
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Maider Badiola-Mateos
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (A.H.); (J.A.d.R.)
| | - José Antonio del Río
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (A.H.); (J.A.d.R.)
| |
Collapse
|
8
|
Sabirzhanov B, Matyas J, Coll-Miro M, Yu LL, Faden AI, Stoica BA, Wu J. Inhibition of microRNA-711 limits angiopoietin-1 and Akt changes, tissue damage, and motor dysfunction after contusive spinal cord injury in mice. Cell Death Dis 2019; 10:839. [PMID: 31685802 PMCID: PMC6828685 DOI: 10.1038/s41419-019-2079-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/09/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) causes neuronal cell death and vascular damage, which contribute to neurological dysfunction. Given that many biochemical changes contribute to such secondary injury, treatment approaches have increasingly focused on combined therapies or use of multi-functional drugs. MicroRNAs (miRs) are small (20-23 nucleotide), non-protein-coding RNAs and can negatively regulate target gene expression at the post-transcriptional level. As individual miRs can potentially modulate expression of multiple relevant proteins after injury, they are attractive candidates as upstream regulators of the secondary SCI progression. In the present study we examined the role of miR-711 modulation after SCI. Levels of miR-711 were increased in injured spinal cord early after SCI, accompanied by rapid downregulation of its target angiopoietin-1 (Ang-1), an endothelial growth factor. Changes of miR-711 were also associated with downregulation of the pro-survival protein Akt (protein kinase B), another target of miR-711, with sequential activation of glycogen synthase kinase 3 and the pro-apoptotic BH3-only molecule PUMA. Central administration of a miR-711 hairpin inhibitor after SCI limited decreases of Ang-1/Akt expression and attenuated apoptotic pathways. Such treatment also reduced neuronal/axonal damage, protected microvasculature and improved motor dysfunction following SCI. In vitro, miR-711 levels were rapidly elevated by neuronal insults, but not by activated microglia and astrocytes. Together, our data suggest that post-traumatic miR-711 elevation contributes to neuronal cell death after SCI, in part by inhibiting Ang-1 and Akt pathways, and may serve as a novel therapeutic target.
Collapse
Affiliation(s)
- Boris Sabirzhanov
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA
| | - Jessica Matyas
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA
| | - Marina Coll-Miro
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA
| | - Laina Lijia Yu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA.,University of Maryland Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), School of Medicine, Baltimore, MD, USA. .,University of Maryland Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA.
| |
Collapse
|
9
|
Yin J, Yin Z, Wang B, Zhu C, Sun C, Liu X, Gong G. Angiopoietin-1 Protects Spinal Cord Ischemia and Reperfusion Injury by Inhibiting Autophagy in Rats. Neurochem Res 2019; 44:2746-2754. [PMID: 31630316 DOI: 10.1007/s11064-019-02893-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/10/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Spinal cord ischemia and reperfusion (SCIR) injury can induce autophagy, which is involved in the survival of neurons. However, whether autophagy plays a neuroprotective or a detrimental role in SCIR injury remains controversial. Angiopoietin-1 (Ang-1), an endothelial growth factor, has been shown to have neuroprotective effects. The present study aimed to explore the neuroprotective mechanisms of Ang-1 in neuronal cells in a rat model of SCIR injury in vivo. Ang-1 protein and rapamycin were injected intrathecally. Basso Beattie Bresnahan (BBB) scoring and hematoxylin and eosin staining were used to assess the degree of SCIR injury. Proteins that reflected the level of autophagy expression, such as Beclin-1 and LC3, were evaluated by western blotting. The results indicated that SCIR injury resulted in loss in lower limb motor function. Ang-1 protein inhibited the expression of Beclin-1 and LC3, which improved the BBB score and alleviated spinal cord injury. In contrast, rapamycin, an autophagy activator, caused the opposite effect. This study provides evidence that Ang-1 plays a neuroprotective role by inhibiting of autophagy expression in SCIR injury. Overall, findings could be useful for the treatment of SCIR injury.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, Lianyungang, 222000, People's Republic of China
| | - Bin Wang
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Chao Zhu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Chao Sun
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China.
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, 211002, Nanjing, People's Republic of China.
| |
Collapse
|
10
|
Yin J, Gong G, Liu X. Angiopoietin: A Novel Neuroprotective/Neurotrophic Agent. Neuroscience 2019; 411:177-184. [PMID: 31152935 DOI: 10.1016/j.neuroscience.2019.05.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 11/29/2022]
Abstract
Angiopoietin (Ang) is an angiogenic factor, but its neuroprotective and neurotrophic effects have recently come to light. Ang exerts neuroprotective effects by inhibiting neuronal apoptosis, protecting the blood-brain/blood-spinal cord barrier, reducing inflammation and promoting neovascularization. In addition, Ang can also promote neural development and neurite outgrowth via activation of the PI3K/Akt signaling pathway and binding to the Tie2 receptor and/or integrin receptor. In addition, Ang and vascular endothelial growth factor (VEGF) are known to interact in blood vessels in the nervous system and the combination of Ang and VEGF can mitigate the negative effects of VEGF, such as inflammation and local edema. These data indicated that Ang is a novel neuroprotective/neurotrophic factor, which may become a new tool for the treatment of nerve injury.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, 211002, China
| | - Xinhui Liu
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.
| |
Collapse
|
11
|
Wang L, Chopp M, Szalad A, Lu X, Lu M, Zhang T, Zhang ZG. Angiopoietin-1/Tie2 signaling pathway contributes to the therapeutic effect of thymosin β4 on diabetic peripheral neuropathy. Neurosci Res 2018; 147:1-8. [PMID: 30326249 DOI: 10.1016/j.neures.2018.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/28/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022]
Abstract
Angiopoietin-1 (Ang1) and its receptor Tie2 regulate vascular function. Our previous study demonstrated that thymosin beta 4 (Tβ4) ameliorates neurological function of diabetic peripheral neuropathy. Mechanisms underlying the therapeutic effect of Tβ4 on diabetic peripheral neuropathy have not been fully investigated. The present in vivo study investigated whether the Ang1/Tie2 signaling pathway is involved in Tβ4-improved neurovascular remodeling in diabetic peripheral neuropathy. Diabetic BKS. Cg-m+/+Leprdb/J (db/db) mice at age 20 weeks were treated with Tβ4 and neutralizing antibody against mouse Tie2 for 4 consecutive weeks. Neurological functional and neurovascular remodeling were measured. Administration of the neutralizing antibody against Tie2 attenuated the therapeutic effect of Tβ4 on improved diabetic peripheral neuropathy as measured by motor and sensory nerve conduction velocity and thermal hypoesthesia compared to diabetic db/db mice treated with Tβ4 only. Histopathological analysis revealed that the neutralizing antibody against Tie2 abolished Tβ4-increased microvascular density in sciatic nerve and intraepidermal nerve fiber density, which were associated with suppression of Tβ4-upregulated occludin expression and Tβ4-reduced protein levels of nuclear factor-κB (NF-κB) and vascular cell adhesion molecule-1 (VCAM1). Our data provide in vivo evidence that the Ang1/Tie2 pathway contributes to the therapeutic effect of Tβ4 on diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA
| | | | - XueRong Lu
- Department of Neurology, Henry Ford Hospital, USA
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Talan Zhang
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | | |
Collapse
|
12
|
Kumar H, Choi H, Jo MJ, Joshi HP, Muttigi M, Bonanomi D, Kim SB, Ban E, Kim A, Lee SH, Kim KT, Sohn S, Zeng X, Han I. Neutrophil elastase inhibition effectively rescued angiopoietin-1 decrease and inhibits glial scar after spinal cord injury. Acta Neuropathol Commun 2018; 6:73. [PMID: 30086801 PMCID: PMC6080383 DOI: 10.1186/s40478-018-0576-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/23/2018] [Indexed: 01/13/2023] Open
Abstract
After spinal cord injury (SCI), neutrophil elastase (NE) released at injury site disrupts vascular endothelium integrity and stabilization. Angiopoietins (ANGPTs) are vascular growth factors that play an important role in vascular stabilization. We hypothesized that neutrophil elastase is one of the key determinants of vascular endothelium disruption/destabilization and affects angiopoietins expression after spinal cord injury. To test this, tubule formation and angiopoietins expression were assessed in endothelial cells exposed to different concentrations of recombinant neutropil elastase. Then, the expression of angiopoietin-1, angiopoietin-2, and neutrophil elastase was determined at 3 h and at 1, 3, 5, 7, 14, 21, and 28 days in a clinically relevant model of moderate compression (35 g for 5 min at T10) spinal cord injury. A dichotomy between the levels of angiopoietin-1 and angiopoietin-2 was observed; thus, we utilized a specific neutrophil elastase inhibitor (sivelestat sodium; 30 mg/kg, i.p., b.i.d.) after spinal cord injury. The expression levels of neutropil elastase and angiopoietin-2 increased, and that of angiopoietin-1 decreased after spinal cord injury in rats. The sivelestat regimen, optimized via a pharmacokinetics study, had potent effects on vascular stabilization by upregulating angiopoietin-1 via the AKT pathway and preventing tight junction protein degradation. Moreover, sivelestat attenuated the levels of inflammatory cytokines and chemokines after spinal cord injury and hence subsequently alleviated secondary damage observed as a reduction in glial scar formation and the promotion of blood vessel formation and stabilization. As a result, hindlimb locomotor function significantly recovered in the sivelestat-treated animals as determined by the Basso, Beattie, and Bresnahan scale and footprint analyses. Furthermore, sivelestat treatment attenuated neuropathic pain as assessed by responses to von Frey filaments after spinal cord injury. Thus, our result suggests that inhibiting neutropil elastase by administration of sivelestat is a promising therapeutic strategy to inhibit glial scar and promote functional recovery by upregulating angiopoietin-1 after spinal cord injury.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Min-Jae Jo
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Manjunatha Muttigi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dario Bonanomi
- Molecular Neurobiology Laboratory, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Sung Bum Kim
- Department of Neurosurgery, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Eunmi Ban
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Aeri Kim
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Neurosurgery, School of Medicine,Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China.
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
13
|
Yin GN, Jin HR, Choi MJ, Limanjaya A, Ghatak K, Minh NN, Ock J, Kwon MH, Song KM, Park HJ, Kim HM, Kwon YG, Ryu JK, Suh JK. Pericyte-Derived Dickkopf2 Regenerates Damaged Penile Neurovasculature Through an Angiopoietin-1-Tie2 Pathway. Diabetes 2018; 67:1149-1161. [PMID: 29559443 DOI: 10.2337/db17-0833] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 03/02/2018] [Indexed: 11/13/2022]
Abstract
Penile erection requires well-coordinated interactions between vascular and nervous systems. Penile neurovascular dysfunction is a major cause of erectile dysfunction (ED) in patients with diabetes, which causes poor response to oral phosphodiesterase-5 inhibitors. Dickkopf2 (DKK2), a Wnt antagonist, is known to promote angiogenesis. Here, using DKK2-Tg mice or DKK2 protein administration, we demonstrate that the overexpression of DKK2 in diabetic mice enhances penile angiogenesis and neural regeneration and restores erectile function. Transcriptome analysis revealed that angiopoietin-1 and angiopoietin-2 are target genes for DKK2. Using an endothelial cell-pericyte coculture system and ex vivo neurite sprouting assay, we found that DKK2-mediated juxtacrine signaling in pericyte-endothelial cell interactions promotes angiogenesis and neural regeneration through an angiopoietin-1-Tie2 pathway, rescuing erectile function in diabetic mice. The dual angiogenic and neurotrophic effects of DKK2, especially as a therapeutic protein, will open new avenues to treating diabetic ED.
Collapse
MESH Headings
- Adult
- Angiopoietin-1/agonists
- Angiopoietin-1/genetics
- Angiopoietin-1/metabolism
- Animals
- Cell Line, Tumor
- Cells, Cultured
- Coculture Techniques
- Crosses, Genetic
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetic Angiopathies/drug therapy
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Nephropathies/drug therapy
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/innervation
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Erectile Dysfunction/complications
- Erectile Dysfunction/drug therapy
- Erectile Dysfunction/metabolism
- Erectile Dysfunction/pathology
- Humans
- Intercellular Signaling Peptides and Proteins/chemistry
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/therapeutic use
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Penis/blood supply
- Penis/innervation
- Penis/metabolism
- Penis/pathology
- Pericytes/drug effects
- Pericytes/metabolism
- Pericytes/pathology
- Receptor, TIE-2/agonists
- Receptor, TIE-2/metabolism
- Wnt Signaling Pathway
- Young Adult
Collapse
Affiliation(s)
- Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hai-Rong Jin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
- Department of Urology, Yuhuangding Hospital, Yantai, Shandong Province, People's Republic of China
| | - Min-Ji Choi
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Anita Limanjaya
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Kalyan Ghatak
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Nguyen Nhat Minh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Mi-Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Kang-Moon Song
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Heon Joo Park
- Hypoxia-Related Disease Research Center, Inha University College of Medicine, Incheon, Republic of Korea
| | - Ho Min Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
- Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
14
|
Conditioned Medium of Bone Marrow-Derived Mesenchymal Stromal Cells as a Therapeutic Approach to Neuropathic Pain: A Preclinical Evaluation. Stem Cells Int 2018. [PMID: 29535781 PMCID: PMC5831939 DOI: 10.1155/2018/8179013] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neuropathic pain is a type of chronic pain caused by injury or dysfunction of the nervous system, without effective therapeutic approaches. Mesenchymal stromal cells (MSCs), through their paracrine action, have great potential in the treatment of this syndrome. In the present study, the therapeutic potential of MSC-derived conditioned medium (CM) was investigated in a mouse model of neuropathic pain induced by partial sciatic nerve ligation (PSL). PSL mice were treated by endovenous route with bone marrow-derived MSCs (1 × 106), CM, or vehicle. Gabapentin was the reference drug. Twelve hours after administration, neuropathic mice treated with CM exhibited an antinociceptive effect that was maintained throughout the evaluation period. MSCs also induced nonreversed antinociception, while gabapentin induced short-lasting antinociception. The levels of IL-1β, TNF-α, and IL-6 were reduced, while IL-10 was enhanced on sciatic nerve and spinal cord by treatment with CM and MSCs. Preliminary analysis of the CM secretome revealed the presence of growth factors and cytokines likely involved in the antinociception. In conclusion, the CM, similar to injection of live cells, produces a powerful and long-lasting antinociceptive effect on neuropathic pain, which is related with modulatory properties on peripheral and central levels of cytokines involved with the maintenance of this syndrome.
Collapse
|
15
|
Yuan Q, Sun L, Yu H, An C. Human microvascular endothelial cell promotes the development of dorsal root ganglion neurons via BDNF pathway in a co-culture system. Biosci Biotechnol Biochem 2017; 81:1335-1342. [PMID: 28394221 DOI: 10.1080/09168451.2017.1313695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Our previous study found that co-culture with human vascular endothelial cells (HMVECs) is beneficial for dorsal root ganglion cells (DRGCs). The goal of the present study is to investigate whether co-culture with HMVECs could promote the development of DRGCs, and whether this effect is induced by the secretion of BDNF by HMVECs. DRGCs were mono-cultured, co-cultured with HMVECs or co-cultured with HMVECs that pre-transfected with BDNF siRNA, the expression of neurite formation and branching factors were determined. The results showed that transfecting with BDNF siRNA inhibited BDNF expression and reduced BDNF secretion. Co-culture with HMVECs increased the expression of Etv4, Etv5, FN-L, FN-M, and GAP-43 in DRGCs that accompanied by the activation of ERK pathway. However, these changes were all reversed by the inhibition of BDNF in HMVECs. In conclusion, our data demonstrate that HMVECs potentiated DRGCs development at least partly by the secretion of BDNF in the co-culture system.
Collapse
Affiliation(s)
- Quan Yuan
- a Department of Orthopedics , Shengjing Hospital of China Medical University , Shenyang , People's Republic of China
| | - Li Sun
- b Department of Nephrology , The First Affiliated Hospital of China Medical University , Shenyang , People's Republic of China
| | - Honghao Yu
- a Department of Orthopedics , Shengjing Hospital of China Medical University , Shenyang , People's Republic of China
| | - Chunhou An
- a Department of Orthopedics , Shengjing Hospital of China Medical University , Shenyang , People's Republic of China
| |
Collapse
|
16
|
Cui WL, Qiu LH, Lian JY, Li JC, Hu J, Liu XL. Cartilage oligomeric matrix protein enhances the vascularization of acellular nerves. Neural Regen Res 2016; 11:512-8. [PMID: 27127495 PMCID: PMC4829021 DOI: 10.4103/1673-5374.179078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vascularization of acellular nerves has been shown to contribute to nerve bridging. In this study, we used a 10-mm sciatic nerve defect model in rats to determine whether cartilage oligomeric matrix protein enhances the vascularization of injured acellular nerves. The rat nerve defects were treated with acellular nerve grafting (control group) alone or acellular nerve grafting combined with intraperitoneal injection of cartilage oligomeric matrix protein (experimental group). As shown through two-dimensional imaging, the vessels began to invade into the acellular nerve graft from both anastomotic ends at day 7 post-operation, and gradually covered the entire graft at day 21. The vascular density, vascular area, and the velocity of revascularization in the experimental group were all higher than those in the control group. These results indicate that cartilage oligomeric matrix protein enhances the vascularization of acellular nerves.
Collapse
Affiliation(s)
- Wei-Ling Cui
- Department of Endocrinology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Long-Hai Qiu
- Department of Orthopaedics and Microsurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jia-Yan Lian
- Department of Endocrinology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jia-Chun Li
- Department of Orthopaedics and Microsurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jun Hu
- Department of Orthopaedics and Microsurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiao-Lin Liu
- Department of Orthopaedics and Microsurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
17
|
Chen YC, Harrison PW, Kotrschal A, Kolm N, Mank JE, Panula P. Expression change in Angiopoietin-1 underlies change in relative brain size in fish. Proc Biol Sci 2015; 282:20150872. [PMID: 26108626 PMCID: PMC4590489 DOI: 10.1098/rspb.2015.0872] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/26/2015] [Indexed: 12/29/2022] Open
Abstract
Brain size varies substantially across the animal kingdom and is often associated with cognitive ability; however, the genetic architecture underpinning natural variation in these key traits is virtually unknown. In order to identify the genetic architecture and loci underlying variation in brain size, we analysed both coding sequence and expression for all the loci expressed in the telencephalon in replicate populations of guppies (Poecilia reticulata) artificially selected for large and small relative brain size. A single gene, Angiopoietin-1 (Ang-1), a regulator of angiogenesis and suspected driver of neural development, was differentially expressed between large- and small-brain populations. Zebra fish (Danio rerio) morphants showed that mild knock down of Ang-1 produces a small-brained phenotype that could be rescued with Ang-1 mRNA. Translation inhibition of Ang-1 resulted in smaller brains in larvae and increased expression of Notch-1, which regulates differentiation of neural stem cells. In situ analysis of newborn large- and small-brained guppies revealed matching expression patterns of Ang-1 and Notch-1 to those observed in zebrafish larvae. Taken together, our results suggest that the genetic architecture affecting brain size in our population may be surprisingly simple, and Ang-1 may be a potentially important locus in the evolution of vertebrate brain size and cognitive ability.
Collapse
Affiliation(s)
- Yu-Chia Chen
- Neuroscience Center and Institute of Biomedicine, Anatomy, University of Helsinki, Haartmaninkatu 8, Helsinki 00290, Finland
| | - Peter W Harrison
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Alexander Kotrschal
- Department of Ecology and Genetics/Animal Ecology, Uppsala University, Norbyvägen 18D, Uppsala 75236, Sweden Department of Zoology/Ethology, Stockholm University, Svante Arrhenius väg 18B, Stockholm 10691, Sweden
| | - Niclas Kolm
- Department of Ecology and Genetics/Animal Ecology, Uppsala University, Norbyvägen 18D, Uppsala 75236, Sweden Department of Zoology/Ethology, Stockholm University, Svante Arrhenius väg 18B, Stockholm 10691, Sweden
| | - Judith E Mank
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Pertti Panula
- Neuroscience Center and Institute of Biomedicine, Anatomy, University of Helsinki, Haartmaninkatu 8, Helsinki 00290, Finland
| |
Collapse
|
18
|
Coste C, Neirinckx V, Gothot A, Wislet S, Rogister B. Are neural crest stem cells the missing link between hematopoietic and neurogenic niches? Front Cell Neurosci 2015; 9:218. [PMID: 26136659 PMCID: PMC4469833 DOI: 10.3389/fncel.2015.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic niches are defined as cellular and molecular microenvironments that regulate hematopoietic stem cell (HSC) function together with stem cell autonomous mechanisms. Many different cell types have been characterized as contributors to the formation of HSC niches, such as osteoblasts, endothelial cells, Schwann cells, and mesenchymal progenitors. These mesenchymal progenitors have themselves been classified as CXC chemokine ligand (CXCL) 12-abundant reticular (CAR) cells, stem cell factor expressing cells, or nestin-positive mesenchymal stem cells (MSCs), which have been recently identified as neural crest-derived cells (NCSCs). Together, these cells are spatially associated with HSCs and believed to provide appropriate microenvironments for HSC self-renewal, differentiation, mobilization and hibernation both by cell-cell contact and soluble factors. Interestingly, it appears that regulatory pathways governing the hematopoietic niche homeostasis are operating in the neurogenic niche as well. Therefore, this review paper aims to compare both the regulation of hematopoietic and neurogenic niches, in order to highlight the role of NCSCs and nervous system components in the development and the regulation of the hematopoietic system.
Collapse
Affiliation(s)
- Cécile Coste
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Virginie Neirinckx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - André Gothot
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cardiovascular Sciences, University of Liège Liège, Belgium ; Hematology Department, University Hospital Liège, Belgium
| | - Sabine Wislet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Bernard Rogister
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium ; Groupe Interdisciplinaire de Génoprotéomique Appliquée-Development, Stem Cells and Regenerative Medicine, University of Liège Liège, Belgium ; Neurology Department, University Hospital Liège, Belgium
| |
Collapse
|
19
|
Cartilage Oligomeric Matrix Protein Angiopoeitin-1 Provides Benefits During Nerve Regeneration In Vivo and In Vitro. Ann Biomed Eng 2015; 43:2924-40. [PMID: 26014362 PMCID: PMC4623068 DOI: 10.1007/s10439-015-1342-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 05/16/2015] [Indexed: 10/28/2022]
Abstract
Our group pioneered the study of nerve regeneration in China and has successfully developed human "acellular nerve grafts (ACNGs)". However, our clinical studies revealed that the effects of ACNGs for long and large nerve defects are far from satisfactory. To improve the efficacy of ACNGs, we combined Cartilage oligomeric matrix protein angiopoietin-1 (COMP-Ang1) with ACNGs in rat sciatic nerve injury models and observed the outcomes via angiographic, morphological, and functional analyses. Co-cultures of endothelial cells (ECs) and dorsal root ganglion neurons (DRGs) were also used to characterize the relationship between neovascularization and nerve regeneration. The results showed significant improvements in early neovascularization, nerve regeneration, and functional outcomes in vivo in the ACNG + COMP-Ang1 group. In vitro, neurite length, and density as well as the expression levels of neurofilament 68 (NF68) and phosphorylated-Tie-2 (p-Tie-2) significantly increased when ECs were co-cultured with DRGs using COMP-Ang1. p-Tie-2 expression dramatically decreased after treatment with a Tie-2 kinase inhibitor (S157701), which consequently decreased the level of NF68. COMP-Ang1 can be concluded to promote early neovascularization followed by brisk nerve regeneration, and the mechanism of this regeneration may involve the modulation of the p-Tie-2 and Tie-2 receptors on ECs. These findings demonstrate that ACNGs can be modified using COMP-Ang1 to improve their efficacy in repairing peripheral nerve defects in clinical trials.
Collapse
|
20
|
Wang L, Chopp M, Szalad A, Jia L, Lu X, Lu M, Zhang L, Zhang Y, Zhang R, Zhang ZG. Sildenafil ameliorates long term peripheral neuropathy in type II diabetic mice. PLoS One 2015; 10:e0118134. [PMID: 25689401 PMCID: PMC4331563 DOI: 10.1371/journal.pone.0118134] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/07/2015] [Indexed: 12/22/2022] Open
Abstract
Diabetic peripheral neuropathy is a common complication of long-standing diabetes mellitus. To mimic clinical trials in which patients with diabetes enrolled have advanced peripheral neuropathy, we investigated the effect of sildenafil, a specific inhibitor of phosphodiesterase type 5 enzyme, on long term peripheral neuropathy in middle aged male mice with type II diabetes. Treatment of diabetic mice (BKS.Cg-m+/+Leprdb/J, db/db) at age 36 weeks with sildenafil significantly increased functional blood vessels and regional blood flow in the sciatic nerve, concurrently with augmentation of intra-epidermal nerve fiber density in the skin and myelinated axons in the sciatic nerve. Functional analysis showed that the sildenafil treatment considerably improved motor and sensory conduction velocities in the sciatic nerve and peripheral thermal stimulus sensitivity compared with the saline treatment. In vitro studies showed that mouse dermal endothelial cells (MDE) cultured under high glucose levels exhibited significant down regulation of angiopoietin 1 (Ang1) expression and reduction of capillary-like tube formation, which were completely reversed by sildenafil. In addition, incubation of dorsal root ganglia (DRG) neurons with conditioned medium harvested from MDE under high glucose levels suppressed neurite outgrowth, where as conditional medium harvested from MDE treated with sildenafil under high glucose levels did not inhibit neurite outgrowth of DRG neurons. Moreover, blockage of the Ang1 receptor, Tie2, with a neutralized antibody against Tie2 abolished the beneficial effect of sildenafil on tube formation and neurite outgrowth. Collectively, our data indicate that sildenafil has a therapeutic effect on long term peripheral neuropathy of middle aged diabetic mice and that improvement of neurovascular dysfunction by sildenafil likely contributes to the amelioration of nerve function. The Ang1/Tie2 signaling pathway may play an important role in these restorative processes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
- * E-mail:
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, 48309, United States of America
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - LongFei Jia
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - XueRong Lu
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - Mei Lu
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - RuiLan Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, Michigan, 48202, United States of America
| |
Collapse
|
21
|
Wang L, Chopp M, Jia L, Lu X, Szalad A, Zhang Y, Zhang R, Zhang ZG. Therapeutic Benefit of Extended Thymosin β4 Treatment Is Independent of Blood Glucose Level in Mice with Diabetic Peripheral Neuropathy. J Diabetes Res 2015; 2015:173656. [PMID: 25945352 PMCID: PMC4405294 DOI: 10.1155/2015/173656] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/06/2015] [Indexed: 11/17/2022] Open
Abstract
Peripheral neuropathy is a chronic complication of diabetes mellitus. To investigated the efficacy and safety of the extended treatment of diabetic peripheral neuropathy with thymosin β4 (Tβ4), male diabetic mice (db/db) at the age of 24 weeks were treated with Tβ4 or saline for 16 consecutive weeks. Treatment of diabetic mice with Tβ4 significantly improved motor (MCV) and sensory (SCV) conduction velocity in the sciatic nerve and the thermal and mechanical latency. However, Tβ4 treatment did not significantly alter blood glucose levels. Treatment with Tβ4 significantly increased intraepidermal nerve fiber density. Furthermore, Tβ4 counteracted the diabetes-induced axon diameter and myelin thickness reductions and the g-ratio increase in sciatic nerve. In vitro, compared with dorsal root ganglia (DRG) neurons derived from nondiabetic mice, DRG neurons derived from diabetic mice exhibited significantly decreased neurite outgrowth, whereas Tβ4 promoted neurite growth in these diabetic DRG neurons. Blockage of the Ang1/Tie2 signaling pathway with a neutralized antibody against Tie2 abolished Tβ4-increased neurite outgrowth. Our data demonstrate that extended Tβ4 treatment ameliorates diabetic-induced axonal degeneration and demyelination, which likely contribute to therapeutic effect of Tβ4 on diabetic neuropathy. The Ang1/Tie2 pathway may mediate Tβ4-induced axonal remodeling.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
- *Lei Wang:
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Longfei Jia
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Xuerong Lu
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - RuiLan Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, 2799 W. Grand Boulevard, Detroit, MI 48202, USA
| |
Collapse
|
22
|
Shen J, Zhu Y, Yu H, Fan ZX, Xiao F, Wu P, Zhang QH, Xiong XX, Pan JW, Zhan RY. Buyang Huanwu decoction increases angiopoietin-1 expression and promotes angiogenesis and functional outcome after focal cerebral ischemia. J Zhejiang Univ Sci B 2014; 15:272-80. [PMID: 24599691 DOI: 10.1631/jzus.b1300166] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Buyang Huanwu decoction (BYHWD), a traditional Chinese herbal prescription, has been widely used clinically to treat stroke in China for hundreds of years; however, the mechanisms of this drug for stroke treatment are still unclear. This study aims to observe the cerebral angiogenesis effects of BYHWD on chronic brain injury after focal cerebral ischemia in rats and to explore its possible mechanisms. The ischemia was induced by occlusion of the right middle cerebral artery for 90 min. BYHWD (12.5 and 25.0 g/(kg ∙ d), equivalent to the dry weight of the raw materials) was orally administered twice a day beginning 2 h after surgery. BYHWD significantly attenuated the neurological dysfunction, infarct volume, and brain atrophy after ischemia. There was a significant increase in the microvessel density, as assessed by immunofluorescence CD31, and a significant increase in angiopoietin-1 (Ang-1) in the penumbra areas of the rats was shown by immunohistochemical staining and Western blotting. The results indicate that the neurorestorative effects of BYHWD are associated with angiogenesis and the enhancement of the expressions of Ang-1 on chronic brain injury after focal cerebral ischemia.
Collapse
Affiliation(s)
- Jian Shen
- Brain Medicine Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China; Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China; Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Signaling Network Map of Endothelial TEK Tyrosine Kinase. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:173026. [PMID: 25371820 PMCID: PMC4211299 DOI: 10.1155/2014/173026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 09/15/2014] [Indexed: 11/28/2022]
Abstract
TEK tyrosine kinase is primarily expressed on endothelial cells and is most commonly referred to as TIE2. TIE2 is a receptor tyrosine kinase modulated by its ligands, angiopoietins, to regulate the development and remodeling of vascular system. It is also one of the critical pathways associated with tumor angiogenesis and familial venous malformations. Apart from the vascular system, TIE2 signaling is also associated with postnatal hematopoiesis. Despite the involvement of TIE2-angiopoietin system in several diseases, the downstream molecular events of TIE2-angiopoietin signaling are not reported in any pathway repository. Therefore, carrying out a detailed review of published literature, we have documented molecular signaling events mediated by TIE2 in response to angiopoietins and developed a network map of TIE2 signaling. The pathway information is freely available to the scientific community through NetPath, a manually curated resource of signaling pathways. We hope that this pathway resource will provide an in-depth view of TIE2-angiopoietin signaling and will lead to identification of potential therapeutic targets for TIE2-angiopoietin associated disorders.
Collapse
|
24
|
Yuan Q, Li JJ, An CH, Sun L. Biological characteristics of rat dorsal root ganglion cell and human vascular endothelial cell in mono- and co-culture. Mol Biol Rep 2014; 41:6949-56. [PMID: 25028268 DOI: 10.1007/s11033-014-3581-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/01/2014] [Indexed: 11/29/2022]
Abstract
This study aimed to evaluate the biological activity of rat dorsal root ganglion cell (DRGC) and human vascular endothelial cell (HMVEC) in mono- and co-culture. Expression levels of vascular endothelial growth factor (VEGF) and nerve growth factor (NGF) mRNA were measured by quantitative real-time RT-PCR (qRT-PCR). Western blot analysis was used to identify VEGF and NGF protein expressions. Cell injury was assessed by measuring cell viability with methylthiazol tetrazolium (MTT) assay. The results showed that VEGF and NGF mRNA levels in the HMVEC+DRGC group were significantly higher than those in the DRGC and HMVEC groups (all p < 0.05). There were also greater increases in both VEGF and NGF protein expressions in the HMVEC+DRGC group than those in the DRGC and HMVEC groups (all p < 0.05). The results of MTT analysis revealed significant differences in cell viability among the HMVEC+DRGC group and the DRGC and HMVEC groups (all p < 0.05). In summary, our findings provide evidence that DRGC and HMVEC in co-culture may exhibit greater biological activity than DRGC in mono-culture.
Collapse
Affiliation(s)
- Quan Yuan
- Department of Orthopedics, Shengjing Hospital, China Medical University, San Hao Street No. 36, Heping District, Shenyang, 110004, People's Republic of China,
| | | | | | | |
Collapse
|
25
|
The angiopoietin:Tie 2 interaction: a potential target for future therapies in human vascular disease. Cytokine Growth Factor Rev 2013; 24:579-92. [PMID: 23838360 DOI: 10.1016/j.cytogfr.2013.05.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 01/06/2023]
Abstract
Angiopoietin-1 and -2 are endogenous ligands for the vascular endothelial receptor tyrosine kinase Tie2. Signalling by angiopoietin-1 promotes vascular endothelial cell survival and the sprouting and reorganisation of blood vessels, as well as inhibiting activation of the vascular endothelial barrier to reduce leakage and leucocyte migration into tissues. Angiopoietin-2 generally has an opposing action, and is released naturally at times of vascular growth and inflammation. There is a significant body of emerging evidence that promoting the actions of angiopoietin-1 through Tie2 is of benefit in pathologies of vascular activation, such as sepsis, stroke, diabetic retinopathy and asthma. Similarly, methods to inhibit the actions of angiopoietin-2 are emerging and have been demonstrated to be of preclinical and clinical benefit in reducing tumour angiogenesis. Here the author reviews the evidence for potential benefits of modulation of the interaction of angiopoietins with Tie2, and the potential applications. Additionally, methods for delivery of the complex protein angiopoietin-1 are discussed, as well as potentially deleterious consequences of administering angiopoietin-1.
Collapse
|
26
|
Prapansilp P, Medana I, Mai NTH, Day NPJ, Phu NH, Yeo TW, Hien TT, White NJ, Anstey NM, Turner GDH. A clinicopathological correlation of the expression of the angiopoietin-Tie-2 receptor pathway in the brain of adults with Plasmodium falciparum malaria. Malar J 2013; 12:50. [PMID: 23383853 PMCID: PMC3570345 DOI: 10.1186/1475-2875-12-50] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/14/2013] [Indexed: 02/08/2023] Open
Abstract
Background Plasma angiopoietin (Ang)-2 is associated with disease severity and mortality in adults and children with falciparum malaria. However the mechanism of action of the angiopoietins in fatal malaria is unclear. This study aimed to determine whether the expression of Ang-1 and Ang-2 and their receptor Tie-2 in cerebral endothelial or parenchymal cells was specific to cerebral malaria (CM), correlated with coma or other severe clinical features, and whether plasma and CSF levels of these markers correlated with the clinical and neuropathological features of severe and fatal malaria in Vietnamese adults. Methods Immunohistochemistry was performed for Ang-1, Ang-2 and Tie-2 on post-mortem brain tissue from fatal malaria cases and controls. Quantitative ELISA for plasma and cerebrospinal fluid levels of Ang-1, Ang-2 and Tie-2 was done to compare fatal cases with surviving patients from the same study. Results Immunohistochemistry revealed significant differences in expression in endothelial and parenchymal cells compared to controls. However there was no significant difference in expression of these markers on endothelial cells, astroglial cells or neurons between CM and non-cerebral malaria cases. Immunostaining of Ang-1, Ang-2 and Tie-2 was also not associated with Plasmodium falciparum-infected erythrocyte sequestration in the brain. However Ang-1 and Ang-2 expression in neurons was significantly correlated with the incidence of microscopic haemorrhages. Plasma levels of Ang-2 and Ang-2/Ang-1 ratio were associated with the number of severe malaria complications and were significant and independent predictors of metabolic acidosis and fatal outcome. Conclusions The independent prognostic significance of Ang-2 and the Ang-2/Ang-1 ratio in severe malaria was confirmed, although immunohistochemistry in fatal cases did not reveal increased expression on brain endothelium in cerebral versus non-cerebral cases. Activation of the Ang-Tie-2 pathway in severe malaria is therefore related to acidosis, number of severity criteria and outcome, but is not a specific event in the brain during cerebral malaria.
Collapse
|
27
|
Cui X, Chopp M, Zacharek A, Cui Y, Roberts C, Chen J. The neurorestorative benefit of GW3965 treatment of stroke in mice. Stroke 2012. [PMID: 23204055 DOI: 10.1161/strokeaha.112.677682] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE GW3965, a synthetic liver X receptor agonist, elevates high-density lipoprotein cholesterol and has antiatherosclerosis and anti-inflammation properties. We tested the hypothesis that GW3965 treatment of stroke increases vascular remodeling, promotes synaptic protein expression and axonal growth in the ischemic brain, and improves functional outcome in mice. METHODS Mice were subjected to transient middle cerebral artery occlusion and treated without or with different doses of GW3965 (5, 10, or 20 mg/kg) starting 24 hours after middle cerebral artery occlusion daily for 14 days. Neurological functional tests, blood high-density lipoprotein cholesterol measurement, and immunostaining were performed. Mouse brain endothelial cells, primary cultured artery explants, and primary cortical neurons cultures were also used in vitro. RESULTS GW3965 treatment of stroke significantly increased blood high-density lipoprotein cholesterol level, synaptic protein expression, axonal density, angiogenesis and arteriogenesis, and Angiopoietin1, Tie2, and occludin expression in the ischemic brain and improved functional outcome compared with middle cerebral artery occlusion control animals (n=10; P<0.05). In vitro, GW3965 and high-density lipoprotein cholesterol also significantly increased capillary-like tube formation and artery explant cell migration as well as neurite outgrowth. Inhibition of Angiopoietin-1 attenuated GW3965-induced tube-formation, artery cell migration, and neurite outgrowth (n=6 per group; P<0.05). CONCLUSIONS These data indicate, for the first time, that GW3965 promotes synaptic protein expression and axonal growth and increases vascular remodeling, which may contribute to improvement of functional outcome after stroke. Increasing Angiopoietin-1/Tie2 signaling activity may play an important role in GW3965-induced brain plasticity and neurological recovery from stroke.
Collapse
Affiliation(s)
- Xu Cui
- Neurology Research, E&R Bldg., Room #3091, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | | | | | | | | | | |
Collapse
|
28
|
Kosacka J, Nowicki M, Klöting N, Kern M, Stumvoll M, Bechmann I, Serke H, Blüher M. COMP-angiopoietin-1 recovers molecular biomarkers of neuropathy and improves vascularisation in sciatic nerve of ob/ob mice. PLoS One 2012; 7:e32881. [PMID: 22412941 PMCID: PMC3295786 DOI: 10.1371/journal.pone.0032881] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 02/05/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Leptin-deficient ob/ob mice are a model of type 2 diabetes induced peripheral neuropathy. Ob/ob mice exhibit obesity, insulin resistance, hyperglycaemia, and alterations of peripheral nerve fibres and endoneural microvessels. Here we test the hypothesis that cartilage oligomeric matrix protein (COMP)-Ang-1, a soluble and stabile form of Ang-1 which promotes angiogenesis and nerve growth, improves regeneration of nerve fibres and endoneural microvessels in ob/ob mice. METHODS AND FINDINGS COMP-Ang-1 (100 ng/ml) or NaCl were intraperitoneally (i.p.) injected into male (N = 184), 3-month old, ob/ob or ob/+ mice for 7 and 21 days. We measured expression of Nf68, GAP43, Cx32, Cx26, Cx43, and TNFα in sciatic nerves using Western blot analysis. To investigate the inflammation in sciatic nerves, numbers of macrophages and T-cells were counted after immunofluorescence staining. In ultrathin section, number of myelinated/non-mylinated nerve fibers, g-ratio, the thickness of Schwann cell basal lamina and microvessel endothelium were investigated. Endoneural microvessels were reconstructed with intracardial FITC injection. Treatment with COMP-Ang-1 over 21 days significantly reduced fasting blood glucose and plasma cholesterol concentrations compared to saline treated ob/ob mice. In addition, COMP-Ang-1 treatment: 1) up-regulated expression of Nf68 and GAP43; 2) improved expression of gap junction proteins including connexin 32 and 26; 3) suppressed the expression of TNFα and Cx43 and 4) led to decreased macrophage and T-cell infiltration in sciatic nerve of ob/ob mice. The significant changes of sciatic nerve ultrastructure were not observed after 21-day long COMP-Ang-1 treatment. COMP-Ang-1 treated ob/ob mice displayed regeneration of small-diameter endoneural microvessels. Effects of COMP-Ang-1 corresponded to increased phosphorylation of Akt and p38 MAPK upon Tie-2 receptor. CONCLUSIONS COMP-Ang-1 recovers molecular biomarkers of neuropathy, promotes angiogenesis and suppresses inflammation in sciatic nerves of ob/ob mice suggesting COMP-Ang-1 as novel treatment option to improve morphologic and protein expression changes associated with diabetic neuropathy.
Collapse
Affiliation(s)
- Joanna Kosacka
- Department of Medicine, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Yan T, Chopp M, Ye X, Liu Z, Zacharek A, Cui Y, Roberts C, Buller B, Chen J. Niaspan increases axonal remodeling after stroke in type 1 diabetes rats. Neurobiol Dis 2012; 46:157-64. [PMID: 22266016 DOI: 10.1016/j.nbd.2012.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/19/2011] [Accepted: 01/03/2012] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVE We investigated axonal plasticity in the bilateral motor cortices and the long term therapeutic effect of Niaspan on axonal remodeling after stroke in type-1 diabetic (T1DM) rats. EXPERIMENTAL APPROACHES T1DM was induced in young adult male Wistar rats via injection of streptozotocin. T1DM rats were subjected to 2h transient middle cerebral artery occlusion (MCAo) and were treated with 40 mg/kg Niaspan or saline starting 24 h after MCAo and daily for 28 days. Anterograde tracing using biotinylated dextran amine (BDA) injected into the contralateral motor cortex was performed to assess axonal sprouting in the ipsilateral motor cortex area. Functional outcome, SMI-31 (a pan-axonal microfilament marker), Bielschowsky silver and synaptophysin expression were measured. In vitro studies using primary cortical neuron (PCN) cultures and in vivo BDA injection into the brain to anterogradely label axons and terminals were employed. RESULTS Niaspan treatment of stroke in T1DM-MCAo rats significantly improved functional outcome after stroke and increased SMI-31, Bielschowsky silver and synaptophysin expression in the ischemic brain compared to saline treated T1DM-MCAo rats (p<0.05). Using BDA to anterograde label axons and terminals, Niaspan treatment significantly increased axonal density in ipsilateral motor cortex in T1DM-MCAo rats (p<0.05, n=7/group). Niacin treatment of PCN significantly increased Ang1 expression under high glucose condition. Niacin and Ang1 significantly increased neurite outgrowth, and anti-Ang1 antibody marginally attenuated Niacin induced neurite outgrowth (p=0.06, n=6/group) in cultured PCN under high glucose condition. CONCLUSION Niaspan treatment increased ischemic brain Ang1 expression and promoted axonal remodeling in the ischemic brain as well as improved functional outcome after stroke. Ang1 may partially contribute to Niaspan-induced axonal remodeling after stroke in T1DM-rats.
Collapse
Affiliation(s)
- Tao Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Angiogenesis is Regulated by Angiopoietins During Experimental Autoimmune Encephalomyelitis and is Indirectly Related to Vascular Permeability. J Neuropathol Exp Neurol 2011; 70:1107-23. [DOI: 10.1097/nen.0b013e31823a8b6a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
31
|
Ward NL, Hatala DA, Wolfram JA, Knutsen DA, Loyd CM. Cutaneous manipulation of vascular growth factors leads to alterations in immunocytes, blood vessels and nerves: Evidence for a cutaneous neurovascular unit. J Dermatol Sci 2010; 61:14-22. [PMID: 21129919 DOI: 10.1016/j.jdermsci.2010.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/26/2010] [Accepted: 11/08/2010] [Indexed: 01/11/2023]
Abstract
BACKGROUND Skin cells produce soluble factors which influence keratinocyte proliferation, angiogenesis, nerve innervation and immunocyte response. OBJECTIVE To test the hypothesis that epidermal-dermal interactions influence neural outgrowth, vascular survival, immunocyte recruitment and keratinocyte proliferation. METHODS We genetically manipulated the epidermis to express excess vascular endothelial growth factor (VEGF) and/or angiopoietin-1 (Ang1) and then examined the epidermal and dermal phenotypes. We compared these findings with those occurring following overexpression of the Ang1 receptor Tie2 in endothelial cells or keratinocytes. RESULTS Keratinocyte-overexpression of Ang1 resulted in increased epidermal thickness compared to control littermates. Keratinocyte-specific overexpression of Ang1 or VEGF increased dermal angiogenesis compared to control animals and combined Ang1-VEGF lead to further increases. Cutaneous leukocyte examination revealed increases in CD4(+) T cell infiltration in mice with keratinocyte-specific overexpression of Ang1, VEGF and Ang1-VEGF combined; in contrast only keratinocyte-specific Ang1 overexpression increased cutaneous F4/80(+) macrophage numbers. Interestingly, combined keratinocyte-derived Ang1-VEGF overexpression reduced significantly the number of F4/80(+) and Cd11c(+) cells compared to mice overexpressing epidermal Ang1 alone. Endothelial cell-specific Tie2 overexpression increased dermal angiogenesis but failed to influence the epidermal and immune cell phenotypes. Keratinocyte-specific Tie2 expressing mice had the highest levels of CD4(+), CD8(+) and CD11c(+) cell numbers and acanthosis compared to all animals. Finally, increases in the number of cutaneous nerves were found in all transgenic mice compared to littermate controls. CONCLUSION These findings demonstrate that change to one system (vascular or epidermal) results in change to other cutaneous systems and suggest that individual molecules can exert effects on multiple systems.
Collapse
Affiliation(s)
- Nicole L Ward
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | | | | | |
Collapse
|
32
|
The angiogenic factor angiopoietin-1 is a proneurogenic peptide on subventricular zone stem/progenitor cells. J Neurosci 2010; 30:4573-84. [PMID: 20357108 DOI: 10.1523/jneurosci.5597-09.2010] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In the adult mammalian brain, the subventricular zone (SVZ) hosts stem cells constantly generating new neurons. Angiopoietin-1 (Ang-1) is an endothelial growth factor with a critical role in division, survival, and adhesion of endothelial cells via Tie-2 receptor activity. Expression of Tie-2 in nonendothelial cells, especially neurons and stem cells, suggests that Ang-1 may be involved in neurogenesis. In the present work, we investigated the putative role of Ang-1 on SVZ neurogenesis. Immature cells from SVZ-derived neurospheres express Ang-1 and Tie-2 mRNA, suggesting a role for the Ang-1/Tie-2 system in the neurogenic niche. Moreover, we also found that Tie-2 protein expression is retained on differentiation in neurons and glial cells. Ang-1 triggered proliferation via activation of the ERK1/2 (extracellular signal-regulated kinase 1/2) mitogen-activated protein kinase (MAPK) kinase pathway but did not induce cell death. Accordingly, coincubation with an anti-Tie-2 neutralizing antibody prevented the pro-proliferative effect of Ang-1. Furthermore, Ang-1 increased the number of NeuN (neuronal nuclear protein)-positive neurons in cultures treated for 7 d, as well as the number of functional neurons, as assessed by monitoring [Ca(2+)](i) rises after application of specific stimuli for neurons and immature cells. The proneurogenic effect of Ang-1 is mediated by Tie-2 activation and subsequent mTOR (mammalian target of rapamycin kinase) mobilization. In agreement, neuronal differentiation significantly decreased after exposure to an anti-Tie-2 neutralizing antibody and to rapamycin. Moreover, Ang-1 elicited the activation of the SAPK (stress-activated protein kinase)/JNK (c-Jun N-terminal kinase) MAPK, involved in axonogenesis. Our work shows a proneurogenic effect of Ang-1, highlighting the relevance of blood vessel/stem cell cross talk in health and disease.
Collapse
|
33
|
Gericke MT, Kosacka J, Koch D, Nowicki M, Schröder T, Ricken AM, Nieber K, Spanel-Borowski K. Receptors for NPY and PACAP differ in expression and activity during adipogenesis in the murine 3T3-L1 fibroblast cell line. Br J Pharmacol 2009; 157:620-32. [PMID: 19422400 DOI: 10.1111/j.1476-5381.2009.00164.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuropeptides are involved in the regulation of food intake in the central nervous system, but they might also act on peripheral fat tissue via neuropeptide receptors. EXPERIMENTAL APPROACH We investigated the receptor expression and activity of pituitary adenylate cyclase-activating polypeptide (PACAP) and of neuropeptide Y at the mRNA and protein levels in the 3T3-L1 fibroblast line during differentiation into adipocytes. Intracellular calcium concentration was measured by calcium imaging. KEY RESULTS The PACAP receptors PAC(1) and VPAC(2) as well as the neuropeptide Y(1) receptor were expressed at the mRNA level in fibroblasts, pre-adipocytes and adipocytes. The mRNA profile of the PAC(1) receptor isoforms showed the HOP sequence, whereas the HIP-isoform was present in subconfluent 3T3-L1 fibroblasts only. At the protein level, the mature 3T3-L1 adipocytes produced the PAC(1) and Y(1) receptors; only the PAC(1) receptor showed carbohydrate residues. Both neuropeptides induced an increase of intracellular calcium in mature adipocytes, which was absent in the precursor cells. These changes in calcium were mediated by Y(1) and PAC(1) receptors as demonstrated by the effects of specific receptor agonists and antagonists. CONCLUSIONS AND IMPLICATIONS As the PAC(1)-HOP receptor variant seems to be responsible for PACAP-mediated calcium influx in many cell types, the HOP sequence might also mediate the increase in intracellular calcium in adipocytes. Because a high intracellular calcium level is associated with lipogenesis, peptidergic innervation of adipose tissue might be involved in stress-induced obesity.
Collapse
|
34
|
Chen X, Fu W, Tung CE, Ward NL. Angiopoietin-1 induces neurite outgrowth of PC12 cells in a Tie2-independent, beta1-integrin-dependent manner. Neurosci Res 2009; 64:348-54. [PMID: 19379779 DOI: 10.1016/j.neures.2009.04.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 03/23/2009] [Accepted: 04/07/2009] [Indexed: 01/17/2023]
Abstract
Overexpression of angiopoietin (Ang) 1 in the brain results in increased vascularization and altered neuronal dendrite configuration. We hypothesized that Ang1 acts directly on neurons inducing neurite outgrowth. We stimulated PC12 cells with Ang1 and observed outgrowth levels comparable to nerve growth factor (NGF). Western blotting and RT-PCR demonstrated the absence of the Ang1 receptor, Tie2 and the presence of beta1-integrin. Downstream of beta1-integrin, Ang1 stimulation led to a approximately 2.6 fold increase in focal adhesion kinase (FAK) phosphorylation and no change in the activation of mitogen-activated protein kinase (MAPK) nor c-Jun N-terminal kinase (JNK). Conversely, NGF stimulation had no effect on FAK phosphorylation but led to a approximately 3.1 and approximately 2 fold increase in phosphorylation of MAPK and JNK. Ang1, but not NGF-mediated outgrowth was attenuated following functional inhibition of beta1-integrin and FAK, and Wortmannin inhibited neurite outgrowth mediated by both. Our results suggest that Ang1 induces neurite outgrowth in PC12 cells in a Tie2-independent, beta1-integrin-FAK-PI3K-Akt-dependent manner and that NGF and Ang1 mediate neurite outgrowth via two independent signaling mechanisms.
Collapse
Affiliation(s)
- Xinyu Chen
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
35
|
Guzmán-Lenis MS, Navarro X, Casas C. Selective sigma receptor agonist 2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate (PRE084) promotes neuroprotection and neurite elongation through protein kinase C (PKC) signaling on motoneurons. Neuroscience 2009; 162:31-8. [PMID: 19345724 DOI: 10.1016/j.neuroscience.2009.03.067] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 02/24/2009] [Accepted: 03/12/2009] [Indexed: 01/23/2023]
Abstract
Neuronal loss and interruption of axonal pathways are occurring after spinal cord injury. This is initiated by the mechanical damage and propagated by secondary events that include the fast rise of glutamate concentration and the subsequent over-activation of glutamate receptors, triggering noxious processes to the cell. Excitotoxic processes are also observed in degenerative diseases that involve motoneuron loss. Sigma-1 receptors (Sig-1Rs) are expressed in the CNS and their ligands have been shown to prevent neuronal death associated to glutamate toxicity. In the present study, we used organotypic cultures of spinal cord slices and dorsal root ganglia (DRG) explants from 7-8 days old postnatal rats to assess whether the agonist of the Sig-1R, 2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate (PRE084), protects the spinal cord against glutamate excitotoxicity and promotes neurite elongation. The results showed that PRE084 exerted a bell-shape dose-dependent protective response of the motoneurons, with a significant neuroprotection obtained with 10 microM PRE084. PRE084 also caused an increase in the length of neurites in both motoneurons and neurons in DRG explants. Both effects were abrogated with the addition of BD 1063, an antagonist of Sig-1R, and the use of chelerythrine, a protein kinase C (PKC) pan-inhibitor indicating that PKC is implicated in the observed effects. These results suggest the use of PRE084 as a neuroprotective agent for spinal cord damage.
Collapse
Affiliation(s)
- M-S Guzmán-Lenis
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | |
Collapse
|
36
|
Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol 2009; 10:165-77. [PMID: 19234476 DOI: 10.1038/nrm2639] [Citation(s) in RCA: 1036] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Angiogenesis, the growth of blood vessels, is a fundamental biological process that controls embryonic development and is also involved in numerous life-threatening human diseases. Much work in the field of angiogenesis research has centred on the vascular endothelial growth factor (VEGF)-VEGF receptor system. The Tie receptors and their angiopoietin (Ang) ligands have been identified as the second vascular tissue-specific receptor Tyr kinase system. Ang-Tie signalling is essential during embryonic vessel assembly and maturation, and functions as a key regulator of adult vascular homeostasis. The structural characteristics and the spatio-temporal regulation of the expression of receptors and ligands provide unique insights into the functions of this vascular signalling system.
Collapse
|
37
|
Bai Y, Cui M, Meng Z, Shen L, He Q, Zhang X, Chen F, Xiao J. Ectopic expression of angiopoietin-1 promotes neuronal differentiation in neural progenitor cells through the Akt pathway. Biochem Biophys Res Commun 2008; 378:296-301. [PMID: 19028450 DOI: 10.1016/j.bbrc.2008.11.052] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2008] [Accepted: 11/14/2008] [Indexed: 12/11/2022]
Abstract
In regions of adult neurogenesis, neural progenitor cells (NPCs) are found in close proximity to blood vessels within a so-called 'vascular niche'. Neurogenesis is linked to angiogenesis via certain growth factors. We propose that angiopoietin-1 (Ang1), which is similar to VEGF, has a unique role in neurogenesis independent of its role in angiogenesis. In this study, primary cultures of NPCs were transduced with recombinant adenoviruses expressing Ang1 and induced to differentiate with dibutyryl cyclic AMP (dbcAMP). Neuronal differentiation was evaluated by quantitative PCR, immunofluorescence microscopy and Western blot analysis. The results show that ectopic expression of Ang1 promotes neuronal differentiation and neurite outgrowth in NPCs, while this effect was blocked by the presence of anti-Tie2 receptor antibody or the PI3-K inhibitor, LY294002. Our results suggest that Ang1, identified originally as an angiogenic factor, can also stimulate in vitro neurogenesis in NPCs through the Akt pathway.
Collapse
Affiliation(s)
- Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian, Beijing 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Gotsch F, Romero R, Kusanovic JP, Chaiworapongsa T, Dombrowski M, Erez O, Than NG, Mazaki-Tovi S, Mittal P, Espinoza J, Hassan SS. Preeclampsia and small-for-gestational age are associated with decreased concentrations of a factor involved in angiogenesis: soluble Tie-2. J Matern Fetal Neonatal Med 2008; 21:389-402. [PMID: 18570117 DOI: 10.1080/14767050802046069] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE An anti-angiogenic state has been described in patients with preeclampsia, small-for-gestational age (SGA) fetuses and fetal death, and changes in the concentration of circulating angiogenic and anti-angiogenic factors can precede the clinical recognition of preeclampsia and SGA by several weeks. Gene deletion studies demonstrate that a selective group of endothelial growth factors are required for vascular development, including members of the vascular endothelial growth factor (VEGF) family, as well as angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), both ligands for the tyrosine kinase endothelial cell receptor Tie-2. These angiogenic factors have been proposed to promote angiogenesis in a coordinated and complementary fashion. Soluble Tie-2 (sTie-2) is the soluble form of the Tie-2 receptor, which is detectable in biological fluids. The purpose of this study was to determine whether patients with preeclampsia and mothers who deliver a SGA neonate have changes in the plasma concentrations of sTie-2. STUDY DESIGN This cross-sectional study included patients in the following groups: (1) non-pregnant women (n = 40), (2) women with normal pregnancies (n = 135), (3) patients with preeclampsia (n = 112), and (4) patients who delivered an SGA neonate (n = 53). Maternal plasma concentrations of sTie-2 were measured by a sensitive immunoassay. Non-parametric statistics were used for analysis. RESULTS (1) The median maternal plasma concentration of sTie-2 was lower in normal pregnant women than in non-pregnant women [median 16.0 ng/mL (range 5.0-71.6) vs. median 20.7 ng/mL (range 10.8-52.4), respectively; p = 0.01)). (2) Plasma sTie-2 concentrations in normal pregnancy changed significantly as a function of gestational age. (3) Patients with preeclampsia and those who delivered SGA neonates had a lower median maternal plasma concentration of sTie-2 than those with a normal pregnancy [preeclampsia: median 14.9 ng/mL (range 4.9-67.3); SGA: median 10.9 ng/mL (range 5.1-29.1); normal pregnancy: median 16.0 ng/mL (range 5.0-71.6); p = 0.048 and p < 0.001, respectively]. (4) Patients with SGA neonates had a lower median plasma concentration of sTie-2 than that of those with preeclampsia [median 10.9 ng/mL (range 5.1-29.1) vs. median 14.9 ng/mL (range 4.9-67.3), respectively; p < 0.001]. (5) Patients with early-onset preeclampsia (<or=34 weeks) had lower concentrations of sTie-2 than women with late-onset preeclampsia (>34 weeks) median of delta values: -0.13 ng/mL (range -0.47-0.58) vs. median of delta values: -0.09 ng/mL (range: -0.60-0.58), respectively; p = 0.043]. In contrast, there were no significant differences in the maternal plasma sTie-2 concentration between women with severe and mild preeclampsia (p = 0.6). CONCLUSION Patients with preeclampsia and those with SGA fetuses have lower median plasma concentrations of soluble Tie-2 than women with normal pregnancies.
Collapse
Affiliation(s)
- Francesca Gotsch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chen J, Cui X, Zacharek A, Chopp M. Increasing Ang1/Tie2 expression by simvastatin treatment induces vascular stabilization and neuroblast migration after stroke. J Cell Mol Med 2008; 13:1348-57. [PMID: 18544044 PMCID: PMC3710660 DOI: 10.1111/j.1582-4934.2008.00380.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, we tested the hypothesis that the Angiopoietin 1 (Ang1)/Tie2 pathway mediates simvastatin-induced vascular integrity and migration of neuroblasts after stroke. Rats were subjected to 2 hrs of middle cerebral artery occlusion (MCAo) and treated, starting 1 day after stroke with or without simvastatin (1 mg/kg, daily) for 7 days. Simvastatin treatment significantly decreased blood–brain barrier (BBB) leakage and concomitantly, increased Ang1, Tie2 and Occludin expression in the ischaemic border (IBZ) compared to the MCAo control group. Simvastatin also significantly increased doublecortin (DCX, a marker of migrating neuroblasts) expression in the IBZ compared to control MCAo rats. DCX was highly expressed around vessels. To further investigate the signalling pathway of simvastatin-induced vascular stabilization and angiogenesis, rat brain microvascular endothelial cell (RBMEC) culture was employed. The data show that simvastatin treatment of RBMEC increased Ang1 and Tie2 gene and protein expression and promoted phosphorylated-Tie2 activity. Simvastatin significantly increased endothelial capillary tube formation, an index of angiogenesis, compared to non-treated control. Inhibition of Ang1 or knockdown of Tie2 gene expression in endothelial cells significantly attenuated simvastatin-induced capillary tube formation. In addition, simvastatin significantly increased subventricular zone (SVZ) explant cell migration compared to non-treatment control. Inhibition of Ang1 significantly attenuated simvastatin-induced SVZ cell migration. Simvastatin treatment of stroke increases Ang1/Tie2 expression and thereby reduces BBB leakage and promotes vascular stabilization. Ang1/Tie2 expression induced by simvastatin treatment promotes neuroblast micro-vascular coupling after stroke.
Collapse
Affiliation(s)
- Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA.
| | | | | | | |
Collapse
|
40
|
Gimble JM, Guilak F, Nuttall ME, Sathishkumar S, Vidal M, Bunnell BA. In vitro Differentiation Potential of Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2008; 35:228-238. [PMID: 21547120 DOI: 10.1159/000124281] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Accepted: 03/07/2008] [Indexed: 12/19/2022]
Abstract
SUMMARY: Mesenchymal stem cells (MSCs) represent a class of multipotent progenitor cells that have been isolated from multiple tissue sites. Of these, adipose tissue and bone marrow offer advantages in terms of access, abundance, and the extent of their documentation in the literature. This review focuses on the in vitro differentiation capability of cells derived from adult human tissue. Multiple, independent studies have demonstrated that MSCs can commit to mesodermal (adipocyte, chondrocyte, hematopoietic support, myocyte, osteoblast, tenocyte), ectodermal (epithelial, glial, neural), and endodermal (hepatocyte, islet cell) lineages. The limitations and promises of these studies in the context of tissue engineering are discussed.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | | | | | | | |
Collapse
|
41
|
Makinde T, Agrawal DK. Intra and extravascular transmembrane signalling of angiopoietin-1-Tie2 receptor in health and disease. J Cell Mol Med 2008; 12:810-28. [PMID: 18266978 PMCID: PMC4401129 DOI: 10.1111/j.1582-4934.2008.00254.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Angiopoietin-1 (Ang-1) is the primary agonist for Tie2 tyrosine kinase receptor (Tie2), and the effect of Ang-1-Tie2 signalling is context-dependent. Deficiency in either Ang-1 or Tie2 protein leads to severe microvascular defects and subsequent embryonic lethality in murine model. Tie2 receptors are expressed in several cell types, including endothelial cells, smooth muscle cells, fibroblasts, epithelial cells, monocytes, neutrophils, eosinophils and glial cells. Ang-1-Tie2 signalling induces a chemotactic effect in smooth muscle cells, neutrophils and eosinophils, and induces differentiation of mesenchymal cells to smooth muscle cells. Additionally, this signalling pathway induces the secretion of serotonin, matrix metalloproteinases (MMPs) and plasmin. Ang-1 inhibits the secretion of tissue inhibitor of matrix metalloproteinase (TIMPs). Aberrant expression and activity of Tie2 in vascular and non-vascular cells may result in the development of rheumatoid arthritis, cancer, hypertension and psoriasis. Ang-1 has an anti-inflammatory effect, when co-localized with vascular endothelial growth factor (VEGF) in the vasculature. Thus, Ang-1 could be potentially important in the therapy of various pathological conditions such as pulmonary hypertension, arteriosclerosis and diabetic retinopathy. In this article, we have summarized and critically reviewed the pathophysiological role of Ang-1-Tie2 signalling pathway.
Collapse
Affiliation(s)
- T Makinde
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | |
Collapse
|
42
|
Abstract
Angiopoietins (ANG-1 and ANG-2) and their TIE-2 receptor tyrosine kinase have wide-ranging effects on tumor malignancy that includes angiogenesis, inflammation, and vascular extravasation. These multifaceted pathways present a valuable opportunity in developing novel inhibition strategies for cancer treatment. However, the regulatory role of ANG-1 and ANG-2 in tumor angiogenesis remains controversial. There is a complex interplay between complementary yet conflicting roles of both the ANGs in shaping the outcome of angiogenesis. Embryonic vascular development suggests that ANG-1 is crucial in engaging interaction between endothelial and perivascular cells. However, recruitment of perivascular cells by ANG-1 has recently been implicated in its antiangiogenic effect on tumor growth. It is becoming clear that TIE-2 signaling may function in a paracrine and autocrine manner directly on tumor cells because the receptor has been increasingly found in tumor cells. In addition, alpha(5)beta(1) and alpha(v)beta(5) integrins were recently recognized as functional receptors for ANG-1 and ANG-2. Therefore, both the ligands may have wide-ranging functions in cellular activities that affect overall tumor development. Collectively, these TIE-2-dependent and TIE-2-independent activities may account for the conflicting findings of ANG-1 and ANG-2 in tumor angiogenesis. These uncertainties have impeded development of a clear strategy to target this important angiogenic pathway. A better understanding of the molecular basis of ANG-1 and ANG-2 activity in the pathophysiologic regulation of angiogenesis may set the stage for novel therapy targeting this pathway.
Collapse
Affiliation(s)
- Winston S N Shim
- Research and Development Unit, National Heart Centre, 17 Third Hospital Avenue, Singapore 168752, Singapore.
| | | | | |
Collapse
|
43
|
Raab S, Plate KH. Different networks, common growth factors: shared growth factors and receptors of the vascular and the nervous system. Acta Neuropathol 2007; 113:607-26. [PMID: 17492293 DOI: 10.1007/s00401-007-0228-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 04/18/2007] [Accepted: 04/18/2007] [Indexed: 01/13/2023]
Abstract
Growth factors and their respective receptors are key regulators during development and for homeostasis of the nervous system. In addition, changes in growth factor function, availability or downstream signaling is involved in many neuropathological disorders like Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, stroke and brain tumours. Research of the recent years revealed that some growth factors, initially discovered as neural growth factors are also affecting blood vessels [e.g. nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF)]. Likewise, vascular growth factors, such as vascular endothelial growth factor (VEGF), which was previously described as an endothelial cell specific mitogen, also affect neural cells. The discovery of shared growth factors affecting the vascular and the nervous system is of relevance for potential therapies of vascular and neurological diseases. This review aims to give an overview about the growing field of common growth factors and receptors within the two different networks.
Collapse
Affiliation(s)
- Sabine Raab
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe-University, Frankfurt/Main, Germany
| | | |
Collapse
|
44
|
Abstract
Angiopoietin-1 (Ang1) has powerful vascular protective effects: suppressing plasma leakage, inhibiting vascular inflammation, and preventing endothelial death. Preclinical studies indicate that Ang1 may be therapeutically useful in a number of situations, including treatment of edema, endotoxemia, and transplant arteriosclerosis. However, the ligand has also been implicated in vessel remodeling, induction of angiogenesis and pulmonary hypertension, indicating that strategies to minimize any deleterious effects while optimizing vessel protection are likely to be needed. This review surveys the published data on vascular protective effects of Ang1 and highlights the therapeutic potential of this ligand, as well as possible limitations to its use. We also consider the data on Ang1 receptors and speculate on how to maximize therapeutic benefit by targeting the Tie receptors.
Collapse
Affiliation(s)
- Nicholas P J Brindle
- Department of Cardiovascular Sciences, University of Leicester, RKCSB, PO Box 65, Leicester, LE2 7LX, UK.
| | | | | |
Collapse
|
45
|
Seifert G, Schilling K, Steinhäuser C. Astrocyte dysfunction in neurological disorders: a molecular perspective. Nat Rev Neurosci 2006; 7:194-206. [PMID: 16495941 DOI: 10.1038/nrn1870] [Citation(s) in RCA: 565] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent work on glial cell physiology has revealed that glial cells, and astrocytes in particular, are much more actively involved in brain information processing than previously thought. This finding has stimulated the view that the active brain should no longer be regarded solely as a network of neuronal contacts, but instead as a circuit of integrated, interactive neurons and glial cells. Consequently, glial cells could also have as yet unexpected roles in the diseased brain. An improved understanding of astrocyte biology and heterogeneity and the involvement of these cells in pathogenesis offers the potential for developing novel strategies to treat neurological disorders.
Collapse
Affiliation(s)
- Gerald Seifert
- Department of Experimental Neurobiology, Clinic of Neurosurgery, University of Bonn, Germany
| | | | | |
Collapse
|
46
|
Kosacka J, Nowicki M, Kacza J, Borlak J, Engele J, Spanel-Borowski K. Adipocyte-derived angiopoietin-1 supports neurite outgrowth and synaptogenesis of sensory neurons. J Neurosci Res 2006; 83:1160-9. [PMID: 16493688 DOI: 10.1002/jnr.20811] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sensory and sympathetic innervation of the white fat tissue (WAT) contributes to lipolysis. In addition, both fiber types adapt in density to weight gain and loss. Because these findings are indicative for a tight control of nerve fiber plasticity by adipokines, we tested whether adipocytes control neurite growth of sensory neurons through angiopoietin-1 (Ang-1). We further considered initial hints that Ang-1-induced neuritogenesis involves transactivation of the high-affinity nerve growth factor (NGF) receptor trkA. Coculturing dorsal root ganglion (DRG) cells with 3T3-L1 adipocytes supported neurite outgrowth. These neurotrophic effects were associated with the increased expression of Ang-1 (presumably in adipocytes) as well as of trkA. The effects were abolished upon inactivating Ang-1 in culture with selective antibodies. Likewise, neurite outgrowth was impaired in the presence of inactivating NGF antibodies as well as upon inhibition of the NGF high-affinity trkA receptor with the antagonist K252a, indicating a tight cooperation of Ang-1 and NGF in the control of neuritogenesis. DRG-adipipocyte cocultures were further used to establish whether sensory neurons would form synaptic contacts with adipocytes. Electron microscopy demonstrated that cultured sensory neurons develop predominantly neuroneuronal synapses but seem to affect adipocytes by synapses en passant. Comparably to the case for neuritogenesis, expression of the presynaptic protein synaptophysin as well of the postsynaptic protein PSD-95 correlated with Ang-1 levels in culture. It is concluded that adipocyte-secreted Ang-1 supports neurite outgrowth, which is involved in synaptogenesis. The novel function of Ang-1 appears to play a physiological role in WAT plasticity.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes/metabolism
- Adipose Tissue/innervation
- Adipose Tissue/metabolism
- Angiopoietin-1/metabolism
- Animals
- Animals, Newborn
- Cell Communication/drug effects
- Cell Communication/physiology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Coculture Techniques
- Disks Large Homolog 4 Protein
- Ganglia, Spinal/growth & development
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/ultrastructure
- Immunohistochemistry
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/metabolism
- Mice
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Nerve Growth Factor/metabolism
- Nerve Growth Factor/pharmacology
- Neurites/drug effects
- Neurites/metabolism
- Neurites/ultrastructure
- Neuronal Plasticity/drug effects
- Neuronal Plasticity/physiology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Neurons, Afferent/ultrastructure
- Rats
- Rats, Inbred WF
- Receptor, trkA/drug effects
- Receptor, trkA/metabolism
- Stem Cells/metabolism
- Synapses/drug effects
- Synapses/metabolism
- Synapses/ultrastructure
- Synaptic Transmission/physiology
- Synaptophysin/metabolism
Collapse
Affiliation(s)
- Joanna Kosacka
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | |
Collapse
|