1
|
Pan Y, Zhang Q, Li C, Li X, Li S, Wang Y, Wang R, Fan J, Tie Y, Zhao X, Gao Y, Wang Y, Sun X. SIRT5 Alleviates Apoptosis of Vascular Endothelial Cells Under Simulated Microgravity via Desuccinylation of ERO1A. Int J Mol Sci 2025; 26:2908. [PMID: 40243486 PMCID: PMC11988372 DOI: 10.3390/ijms26072908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
The adverse effects of weightlessness on the human cardiovascular system greatly hinder the process of long-term and long-distance space exploration. Succinylation is an important type of protein post-translational modification. However, whether succinylation modification is able to play a role in altered vascular endothelial cell function under microgravity or simulated microgravity has not been reported. This study aims to investigate the quantitative global proteome and the changes in lysine succinylation in related proteins, seeking to facilitate a better understanding of the protein post-translational modification in cardiovascular deconditioning under microgravity. LC-MS/MS combined with bioinformatics analysis were used to quantitatively detect the perspectives at the global protein level. Immunoprecipitation and Western blot analysis were conducted to further verify the alterations of related proteins and lysine succinylation. A total of 132 differentially expressed proteins and 164 differentially expressed lysine succinylation sites were identified in human umbilical vein endothelial cells (HUVECs). Bioinformatics analysis indicates that lysine succinylation may play a potential role in energy metabolism. In addition, desuccinylase SIRT5 was downregulated and regulated succinylation modification levels of HUVECs under simulated microgravity. Notably, the overexpression of SIRT5 effectively protected HUVECs from apoptosis induced by simulated microgravity. And the succinylation of Lys396 in ERO1A was significantly increased in HUVECs under simulated microgravity. Mechanistically, the knockdown of SIRT5 was found to induce the apoptosis of HUVECs through the succinylation of Lys396 in ERO1A. These results can provide new ideas for elucidating the molecular mechanism of cardiovascular dysfunction in microgravity environments, and provide key molecular targets for scientific protective measures against microgravity in space.
Collapse
Affiliation(s)
- Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Qian Zhang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
- College of Life Sciences, Yan’an University, Yan’an 716000, China
| | - Chengfei Li
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi’an 710032, China; (C.L.); (X.L.); (J.F.); (X.Z.)
| | - Xi Li
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi’an 710032, China; (C.L.); (X.L.); (J.F.); (X.Z.)
| | - Shuhan Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Yuan Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Ruonan Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Jieyi Fan
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi’an 710032, China; (C.L.); (X.L.); (J.F.); (X.Z.)
| | - Yateng Tie
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Xingcheng Zhao
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi’an 710032, China; (C.L.); (X.L.); (J.F.); (X.Z.)
| | - Yuan Gao
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| | - Yongchun Wang
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi’an 710032, China; (C.L.); (X.L.); (J.F.); (X.Z.)
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.P.); (Q.Z.); (S.L.); (Y.W.); (R.W.); (Y.T.); (Y.G.)
| |
Collapse
|
2
|
Manna OM, Burgio S, Picone D, Carista A, Pitruzzella A, Fucarino A, Bucchieri F. Microgravity and Human Body: Unraveling the Potential Role of Heat-Shock Proteins in Spaceflight and Future Space Missions. BIOLOGY 2024; 13:921. [PMID: 39596876 PMCID: PMC11591694 DOI: 10.3390/biology13110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
In recent years, the increasing number of long-duration space missions has prompted the scientific community to undertake a more comprehensive examination of the impact of microgravity on the human body during spaceflight. This review aims to assess the current knowledge regarding the consequences of exposure to an extreme environment, like microgravity, on the human body, focusing on the role of heat-shock proteins (HSPs). Previous studies have demonstrated that long-term exposure to microgravity during spaceflight can cause various changes in the human body, such as muscle atrophy, changes in muscle fiber composition, cardiovascular function, bone density, and even immune system functions. It has been postulated that heat-shock proteins (HSPs) may play a role in mitigating the harmful effects of microgravity-induced stress. According to past studies, heat-shock proteins (HSPs) are upregulated under simulated microgravity conditions. This upregulation assists in the maintenance of the proper folding and function of other proteins during stressful conditions, thereby safeguarding the physiological systems of organisms from the detrimental effects of microgravity. HSPs could also be used as biomarkers to assess the level of cellular stress in tissues and cells exposed to microgravity. Therefore, modulation of HSPs by drugs and genetic or environmental techniques could prove to be a potential therapeutic strategy to reduce the negative physiological consequences of long-duration spaceflight in astronauts.
Collapse
Affiliation(s)
- Olga Maria Manna
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
| | - Stefano Burgio
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
- Department of Medicine and Surgery, Kore University of Enna, 94100 Enna, Italy
| | - Domiziana Picone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Adelaide Carista
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Alberto Fucarino
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
3
|
Abdelfattah F, Schulz H, Wehland M, Corydon TJ, Sahana J, Kraus A, Krüger M, González-Torres LF, Cortés-Sánchez JL, Wise PM, Mushunuri A, Hemmersbach R, Liemersdorf C, Infanger M, Grimm D. Omics Studies of Specialized Cells and Stem Cells under Microgravity Conditions. Int J Mol Sci 2024; 25:10014. [PMID: 39337501 PMCID: PMC11431953 DOI: 10.3390/ijms251810014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
The primary objective of omics in space with focus on the human organism is to characterize and quantify biological factors that alter structure, morphology, function, and dynamics of human cells exposed to microgravity. This review discusses exciting data regarding genomics, transcriptomics, epigenomics, metabolomics, and proteomics of human cells and individuals in space, as well as cells cultured under simulated microgravity. The NASA Twins Study significantly heightened interest in applying omics technologies and bioinformatics in space and terrestrial environments. Here, we present the available publications in this field with a focus on specialized cells and stem cells exposed to real and simulated microgravity conditions. We summarize current knowledge of the following topics: (i) omics studies on stem cells, (ii) omics studies on benign specialized different cell types of the human organism, (iii) discussing the advantages of this knowledge for space commercialization and exploration, and (iv) summarizing the emerging opportunities for translational regenerative medicine for space travelers and human patients on Earth.
Collapse
Affiliation(s)
- Fatima Abdelfattah
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| | - Armin Kraus
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Luis Fernando González-Torres
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Ashwini Mushunuri
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Ruth Hemmersbach
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Christian Liemersdorf
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| |
Collapse
|
4
|
Yao H, Tang L, Wang D, Pang H, Yang K. F-actin microfilaments affect the LIPUS-promoted osteogenic differentiation of BMSCs through TRPM7. Biotechnol J 2024; 19:e2400310. [PMID: 39212193 DOI: 10.1002/biot.202400310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
The differentiation of bone marrow mesenchymal stem cells (BMSCs) toward osteogenesis can be induced by low-intensity pulsed ultrasound (LIPUS). However, the molecular mechanisms responsible for LIPUS stimulation are unclear. The possible molecular mechanisms by which LIPUS promotes osteogenic differentiation of BMSCs were investigated in this study. The quantification of alkaline phosphatase (ALP) activity, Alizarin Red S staining, ALP staining, and the establishment of a calvarial defect model were used to evaluate osteogenic effects. Immunofluorescence was performed to observe the expression of microfilaments and transient receptor potential melastatin 7 (TRPM7). The levels of F-actin/G-actin and osteogenesis-related proteins under LIPUS alone or LIPUS combined with cytoskeleton interfering drugs (Cytochalasin D [CytoD] or Jasplakinolide [JA]) were assayed by western blot. Quantitative real-time reverse transcription polymerase chain reaction was utilized to measure the expression of Trpm7 mRNA. Moreover, adenoviral Trpm7 knockdown was verified using western blot. The results demonstrated that LIPUS promoted bone formation in vivo. Under osteogenic induction in vitro, the osteogenesis of BMSCs induced by LIPUS was accompanied by the depolymerization and rearrangement of microfilaments and increased levels of TRPM7. By perturbing intracellular actin dynamics, CytoD enhanced the pro-osteogenicity of LIPUS and increased TRPM7 level, while JA inhibited the pro-osteogenicity of LIPUS and reduced TRPM7 level. Additionally, the knockdown of Trpm7 suppressed the osteogenic promotion of BMSCs induced by LIPUS. The transient depolymerization and rearrangement of the cytoskeleton microfilaments mediated by LIPUS can affect TRPM7 expression and subsequently promote the osteogenesis of BMSCs. This study provides further direction for exploring the molecular mechanism of LIPUS, as a mechanical stress, in facilitating the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Huan Yao
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Wang
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Yang
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Wang Y, Li C, Wang R, Zhao X, Pan Y, Zhang Q, Li S, Fan J, Wang Y, Sun X. PIEZO1 Promotes the Migration of Endothelial Cells via Enhancing CXCR4 Expression under Simulated Microgravity. Int J Mol Sci 2024; 25:7254. [PMID: 39000362 PMCID: PMC11242226 DOI: 10.3390/ijms25137254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Exposure to microgravity during spaceflight induces the alterations in endothelial cell function associated with post-flight cardiovascular deconditioning. PIEZO1 is a major mechanosensitive ion channel that regulates endothelial cell function. In this study, we used a two-dimensional clinostat to investigate the expression of PIEZO1 and its regulatory mechanism on human umbilical vein endothelial cells (HUVECs) under simulated microgravity. Utilizing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis, we observed that PIEZO1 expression was significantly increased in response to simulated microgravity. Moreover, we found microgravity promoted endothelial cells migration by increasing expression of PIEZO1. Proteomics analysis highlighted the importance of C-X-C chemokine receptor type 4(CXCR4) as a main target molecule of PIEZO1 in HUVECs. CXCR4 protein level was increased with simulated microgravity and decreased with PIEZO1 knock down. The mechanistic study showed that PIEZO1 enhances CXCR4 expression via Ca2+ influx. In addition, CXCR4 could promote endothelial cell migration under simulated microgravity. Taken together, these results suggest that the upregulation of PIEZO1 in response to simulated microgravity regulates endothelial cell migration due to enhancing CXCR4 expression via Ca2+ influx.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (Y.W.); (C.L.); (R.W.); (X.Z.); (Y.P.); (Q.Z.); (S.L.); (J.F.); (Y.W.)
| |
Collapse
|
6
|
Lv Y, Yu W, Xuan R, Yang Y, Xue X, Ma X. Human Placental Mesenchymal Stem Cells-Exosomes Alleviate Endothelial Barrier Dysfunction via Cytoskeletal Remodeling through hsa-miR-148a-3p/ROCK1 Pathway. Stem Cells Int 2024; 2024:2172632. [PMID: 38681858 PMCID: PMC11055650 DOI: 10.1155/2024/2172632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Background Endothelial barrier disruption of human pulmonary vascular endothelial cells (HPVECs) is an important pathogenic factor for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Mesenchymal stem cells-exosome (MSCs-Exo) represents an ideal carrier for cell-free therapy. The therapeutic implication and underlying mechanism of human placental MSCs-Exo (HPMSCs-Exo) in ALI/ARDS need to be further explored. Materials and Methods HPMSCs-Exo was extracted from HPMSCs and characterized. Then, the therapeutic effects of exosomes were evaluated in ALI mice and HPVECs. RNA-sequencing was applied to reveal the miRNA profile of HPMSCs-Exo and differentially expressed genes (DEGs) in HPMSCs-Exo-pretreated HPVECs. The targets of miRNAs were predicted by bioinformatics methods and correlated to DEGs. Finally, the role of hsa-miR-148a-3p/ROCK1 pathway in HPVECs has been further discussed. Results The results showed that HPMSCs-Exo could downregulate Rho-associated coiled-coil-containing protein kinase 1 (ROCK1), upregulate the expression of zonula occludens-1 (ZO-1) and F-actin, promote HPVECs migration and tube formation, reduce cytoskeletal disorders and cell permeability, and thus improve ALI/ARDS. RNA-sequencing revealed the DEGs were mainly enriched in cell junction, angiogenesis, inflammation, and energy metabolism. HPMSCs-Exo contains multiple miRNAs which are associated with cytoskeletal function; the expression abundance of hsa-miR-148a-3p is the highest. Bioinformatic analysis identified ROCK1 as a target of hsa-miR-148a-3p. The overexpression of hsa-miR-148a-3p in HPMSCs-Exo promoted the migration and tube formation of HPVECs and reduced ROCK1 expression. However, the overexpression of ROCK1 on HPVECs reduced the therapeutic effect of HPMSCs-Exo. Conclusions HPMSCs-Exo represents a protective regimen against endothelial barrier disruption of HPVECs in ALI/ARDS, and the hsa-miR-148a-3p/ROCK1 pathway plays an important role in this therapeutics implication.
Collapse
Affiliation(s)
- Yuzhen Lv
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Wenqin Yu
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Ruiui Xuan
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Yulu Yang
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaolan Xue
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaowei Ma
- Intensive Care Unit, Cardiocerebral Vascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| |
Collapse
|
7
|
Davis T, Tabury K, Zhu S, Angeloni D, Baatout S, Benchoua A, Bereiter-Hahn J, Bottai D, Buchheim JI, Calvaruso M, Carnero-Diaz E, Castiglioni S, Cavalieri D, Ceccarelli G, Choukér A, Cialdai F, Ciofani G, Coppola G, Cusella G, Degl'Innocenti A, Desaphy JF, Frippiat JP, Gelinsky M, Genchi G, Grano M, Grimm D, Guignandon A, Hahn C, Hatton J, Herranz R, Hellweg CE, Iorio CS, Karapantsios T, van Loon JJWA, Lulli M, Maier J, Malda J, Mamaca E, Morbidelli L, van Ombergen A, Osterman A, Ovsianikov A, Pampaloni F, Pavezlorie E, Pereda-Campos V, Przybyla C, Puhl C, Rettberg P, Rizzo AM, Robson-Brown K, Rossi L, Russo G, Salvetti A, Santucci D, Sperl M, Tavella S, Thielemann C, Willaert R, Szewczyk N, Monici M. How are cell and tissue structure and function influenced by gravity and what are the gravity perception mechanisms? NPJ Microgravity 2024; 10:16. [PMID: 38341423 DOI: 10.1038/s41526-024-00357-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Progress in mechanobiology allowed us to better understand the important role of mechanical forces in the regulation of biological processes. Space research in the field of life sciences clearly showed that gravity plays a crucial role in biological processes. The space environment offers the unique opportunity to carry out experiments without gravity, helping us not only to understand the effects of gravitational alterations on biological systems but also the mechanisms underlying mechanoperception and cell/tissue response to mechanical and gravitational stresses. Despite the progress made so far, for future space exploration programs it is necessary to increase our knowledge on the mechanotransduction processes as well as on the molecular mechanisms underlying microgravity-induced cell and tissue alterations. This white paper reports the suggestions and recommendations of the SciSpacE Science Community for the elaboration of the section of the European Space Agency roadmap "Biology in Space and Analogue Environments" focusing on "How are cells and tissues influenced by gravity and what are the gravity perception mechanisms?" The knowledge gaps that prevent the Science Community from fully answering this question and the activities proposed to fill them are discussed.
Collapse
Affiliation(s)
- Trent Davis
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Kevin Tabury
- Laboratory of Radiobiology, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shouan Zhu
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Debora Angeloni
- Institute of Biorobotics, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Sarah Baatout
- Laboratory of Radiobiology, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | | | - Juergen Bereiter-Hahn
- Institute for Cell Biology and Neurobiology, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Daniele Bottai
- Department Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Judith-Irina Buchheim
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Marco Calvaruso
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | - Eugénie Carnero-Diaz
- Institute of Systematics, Evolution, Biodiversity, Sorbonne University, NMNH, CNRS, EPHE, UA, Paris, France
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | | | - Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Alexander Choukér
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Research Division, DSBSC-University of Florence, Florence, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, 56025, Italy
| | - Giuseppe Coppola
- Institute of Applied Science and Intelligent Systems - CNR, Naples, Italy
| | - Gabriella Cusella
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Andrea Degl'Innocenti
- Department of Medical Biotechnologies, University of Siena, Italy and Smart Bio-Interfaces, IIT, Pontedera, PI, Italy
| | - Jean-Francois Desaphy
- Department of Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Jean-Pol Frippiat
- Stress, Immunity, Pathogens Laboratory, SIMPA, Université de Lorraine, Nancy, France
| | - Michael Gelinsky
- Centre for Translational Bone, Joint & Soft Tissue Research, TU Dresden, Dresden, Germany
| | - Giada Genchi
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, 56025, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University Magdeburg, Germany & Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Alain Guignandon
- SAINBIOSE, INSERM U1059, Université Jean Monnet, Saint-Etienne, F-42000, France
| | | | - Jason Hatton
- European Space Agency, ESTEC, Noordwijk, The Netherlands
| | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Christine E Hellweg
- Radiation Biology Department, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | | | | | | | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Jeanette Maier
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht & Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Emina Mamaca
- European and International Affairs Department, Ifremer centre Bretagne, Plouzané, France
| | | | | | - Andreas Osterman
- Max von Pettenkofer Institute, Virology, LMU Munich & DZIF, Partner Site Munich, Munich, Germany
| | - Aleksandr Ovsianikov
- 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria
| | - Francesco Pampaloni
- Buchmann Inst. for Molecular Life Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Elizabeth Pavezlorie
- Ludwig Boltzmann Institute for Traumatology, Research Center in Cooperation with AUVA, Vienna, Austria
| | - Veronica Pereda-Campos
- GSBMS/URU EVOLSAN - Medecine Evolutive, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Cyrille Przybyla
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Palavas les Flots, France
| | - Christopher Puhl
- Space Applications NV/SA for European Space Agency, Noordwijk, The Netherlands
| | - Petra Rettberg
- DLR, Institute of Aerospace Medicine, Research Group Astrobiology, Köln, Germany
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Kate Robson-Brown
- Department of Engineering Mathematics, and Department of Anthropology and Archaeology, University of Bristol, Bristol, UK
| | - Leonardo Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniela Santucci
- Center for Behavioural Sciences and Mental Health, Istituto Superiore Sanità, Rome, Italy
| | | | - Sara Tavella
- IRCCS Ospedale Policlinico San Martino and University of Genoa, DIMES, Genoa, Italy
| | | | - Ronnie Willaert
- Research Group NAMI and NANO, Vrije Universiteit Brussels, Brussels, Belgium
| | - Nathaniel Szewczyk
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Research Division, DSBSC-University of Florence, Florence, Italy.
| |
Collapse
|
8
|
Hiratsuka K, Miyoshi T, Kroll KT, Gupta NR, Valerius MT, Ferrante T, Yamashita M, Lewis JA, Morizane R. Organoid-on-a-chip model of human ARPKD reveals mechanosensing pathomechanisms for drug discovery. SCIENCE ADVANCES 2022; 8:eabq0866. [PMID: 36129975 PMCID: PMC9491724 DOI: 10.1126/sciadv.abq0866] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/03/2022] [Indexed: 05/23/2023]
Abstract
Organoids serve as a novel tool for disease modeling in three-dimensional multicellular contexts. Static organoids, however, lack the requisite biophysical microenvironment such as fluid flow, limiting their ability to faithfully recapitulate disease pathology. Here, we unite organoids with organ-on-a-chip technology to unravel disease pathology and develop therapies for autosomal recessive polycystic kidney disease. PKHD1-mutant organoids-on-a-chip are subjected to flow that induces clinically relevant phenotypes of distal nephron dilatation. Transcriptomics discover 229 signal pathways that are not identified by static models. Mechanosensing molecules, RAC1 and FOS, are identified as potential therapeutic targets and validated by patient kidney samples. On the basis of this insight, we tested two U.S. Food and Drug Administration-approved and one investigational new drugs that target RAC1 and FOS in our organoid-on-a-chip model, which suppressed cyst formation. Our observations highlight the vast potential of organoid-on-a-chip models to elucidate complex disease mechanisms for therapeutic testing and discovery.
Collapse
Affiliation(s)
- Ken Hiratsuka
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tomoya Miyoshi
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Katharina T. Kroll
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Navin R. Gupta
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
| | - M. Todd Valerius
- Harvard Medical School, Boston, MA, USA
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
| | - Thomas Ferrante
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michifumi Yamashita
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer A. Lewis
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
| | - Ryuji Morizane
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
| |
Collapse
|
9
|
Locatelli L, Castiglioni S, Maier JAM. From Cultured Vascular Cells to Vessels: The Cellular and Molecular Basis of Vascular Dysfunction in Space. Front Bioeng Biotechnol 2022; 10:862059. [PMID: 35480977 PMCID: PMC9036997 DOI: 10.3389/fbioe.2022.862059] [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: 01/25/2022] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Life evolved on this planet under the pull of gravity, shielded from radiation by the magnetosphere and shaped by circadian rhythms due to Earth’s rotation on its axis. Once living beings leave such a protective environment, adaptive responses are activated to grant survival. In view of long manned mission out of Earth’s orbit, it is relevant to understand how humans adapt to space and if the responses activated might reveal detrimental in the long run. Here we review present knowledge about the effects on the vessels of various extraterrestrial factors on humans as well as in vivo and in vitro experimental models. It emerges that the vasculature activates complex adaptive responses finalized to supply oxygen and nutrients to all the tissues and to remove metabolic waste and carbon dioxide. Most studies point to oxidative stress and mitochondrial dysfunction as mediators of vascular alterations in space. Unraveling the cellular and molecular mechanisms involved in these adaptive processes might offer hints to design proper and personalized countermeasures to predict a safe future in space.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Jeanette A M Maier
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy.,Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa), Università di Milano, Milan, Italy
| |
Collapse
|
10
|
Locatelli L, Inglebert M, Scrimieri R, Sinha PK, Zuccotti GV, Milani P, Bureau L, Misbah C, Maier JAM. Human endothelial cells in high glucose: New clues from culture in 3D microfluidic chips. FASEB J 2022; 36:e22137. [DOI: 10.1096/fj.202100914r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/12/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Locatelli
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco Università di Milano Milano Italy
| | | | - Roberta Scrimieri
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco Università di Milano Milano Italy
| | | | - Gian Vincenzo Zuccotti
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco Università di Milano Milano Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) Università di Milano Milano Italy
- Dipartimento di Fisica “A. Pontremoli” Università di Milano Milano Italy
| | - Lionel Bureau
- LIPhy, CNRS Université Grenoble‐Alpes Grenoble France
| | | | - Jeanette A. M. Maier
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco Università di Milano Milano Italy
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) Università di Milano Milano Italy
| |
Collapse
|