1
|
Ya J, Pellumbaj J, Hashmat A, Bayraktutan U. The Role of Stem Cells as Therapeutics for Ischaemic Stroke. Cells 2024; 13:112. [PMID: 38247804 PMCID: PMC10814781 DOI: 10.3390/cells13020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Stroke remains one of the leading causes of death and disability worldwide. Current reperfusion treatments for ischaemic stroke are limited due to their narrow therapeutic window in rescuing ischaemic penumbra. Stem cell therapy offers a promising alternative. As a regenerative medicine, stem cells offer a wider range of treatment strategies, including long-term intervention for chronic patients, through the reparation and replacement of injured cells via mechanisms of differentiation and proliferation. The purpose of this review is to evaluate the therapeutic role of stem cells for ischaemic stroke. This paper discusses the pathology during acute, subacute, and chronic phases of cerebral ischaemic injury, highlights the mechanisms involved in mesenchymal, endothelial, haematopoietic, and neural stem cell-mediated cerebrovascular regeneration, and evaluates the pre-clinical and clinical data concerning the safety and efficacy of stem cell-based treatments. The treatment of stroke patients with different types of stem cells appears to be safe and efficacious even at relatively higher concentrations irrespective of the route and timing of administration. The priming or pre-conditioning of cells prior to administration appears to help augment their therapeutic impact. However, larger patient cohorts and later-phase trials are required to consolidate these findings.
Collapse
Affiliation(s)
| | | | | | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neurosciences, Queens Medical Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
2
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
3
|
Burns N, Nijmeh H, Lapel M, Riddle S, Yegutkin GG, Stenmark KR, Gerasimovskaya E. Isolation of vasa vasorum endothelial cells from pulmonary artery adventitia: Implementation to vascular biology research. Microvasc Res 2023; 147:104479. [PMID: 36690271 DOI: 10.1016/j.mvr.2023.104479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Isolated endothelial cells are valuable in vitro model for vascular research. At present, investigation of disease-relevant changes in vascular endothelium at the molecular level requires established endothelial cell cultures, preserving vascular bed-specific phenotypic characteristics. Vasa vasorum (VV) form a microvascular network around large blood vessels, in both the pulmonary and systemic circulations, that are critically important for maintaining the integrity and oxygen supply of the vascular wall. However, despite the pathophysiological significance of the VV, methods for the isolation and culture of vasa vasorum endothelial cells (VVEC) have not yet been reported. In our prior studies, we demonstrated the presence of hypoxia-induced angiogenic expansion of the VV in the pulmonary artery (PA) of neonatal calves; an observation which has been followed by a series of in vitro studies on isolated PA VVEC. Here we present a detailed protocol for reproducible isolation, purification, and culture of PA VVEC. We show these cells to express generic endothelial markers, (vWF, eNOS, VEGFR2, Tie1, and CD31), as well as progenitor markers (CD34 and CD133), bind lectin Lycopersicon Esculentum, and incorporate acetylated low-density lipoproteins labeled with acetylated LDL (DiI-Ac-LDL). qPCR analysis additionally revealed the expression of CD105, VCAM-1, ICAM-1, MCAM, and NCAM. Ultrastructural electron microscopy and immunofluorescence staining demonstrated that VVEC are morphologically characterized by a developed actin and microtubular cytoskeleton, mitochondrial network, abundant intracellular vacuolar/secretory system, and cell-surface filopodia. VVEC exhibit exponential growth in culture and can be mitogenically activated by multiple growth factors. Thus, our protocol provides the opportunity for VVEC isolation from the PA, and potentially from other large vessels, enabling advances in VV research.
Collapse
Affiliation(s)
- Nana Burns
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Hala Nijmeh
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Martin Lapel
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Suzette Riddle
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Kurt R Stenmark
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Evgenia Gerasimovskaya
- Department of Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, CO, United States of America.
| |
Collapse
|
4
|
Miranda MA, Macias-Velasco JF, Schmidt H, Lawson HA. Integrated transcriptomics contrasts fatty acid metabolism with hypoxia response in β-cell subpopulations associated with glycemic control. BMC Genomics 2023; 24:156. [PMID: 36978008 PMCID: PMC10052828 DOI: 10.1186/s12864-023-09232-5] [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: 05/24/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Understanding how heterogeneous β-cell function impacts diabetes is imperative for therapy development. Standard single-cell RNA sequencing analysis illuminates some factors driving heterogeneity, but new strategies are required to enhance information capture. RESULTS We integrate pancreatic islet single-cell and bulk RNA sequencing data to identify β-cell subpopulations based on gene expression and characterize genetic networks associated with β-cell function in obese SM/J mice. We identify β-cell subpopulations associated with basal insulin secretion, hypoxia response, cell polarity, and stress response. Network analysis associates fatty acid metabolism and basal insulin secretion with hyperglycemic-obesity, while expression of Pdyn and hypoxia response is associated with normoglycemic-obesity. CONCLUSIONS By integrating single-cell and bulk islet transcriptomes, our study explores β-cell heterogeneity and identifies novel subpopulations and genetic pathways associated with β-cell function in obesity.
Collapse
Affiliation(s)
- Mario A Miranda
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Juan F Macias-Velasco
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Heather Schmidt
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA
| | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8232, Saint Louis, MO, 63110, USA.
| |
Collapse
|
5
|
CoCl2-Mimicked Endothelial Cell Hypoxia Induces Nucleotide Depletion and Functional Impairment That Is Reversed by Nucleotide Precursors. Biomedicines 2022; 10:biomedicines10071540. [PMID: 35884844 PMCID: PMC9313011 DOI: 10.3390/biomedicines10071540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic hypoxia drives vascular dysfunction by various mechanisms, including changes in mitochondrial respiration. Although endothelial cells (ECs) rely predominantly on glycolysis, hypoxia is known to alter oxidative phosphorylation, promote oxidative stress and induce dysfunction in ECs. Our work aimed to analyze the effects of prolonged treatment with hypoxia-mimetic agent CoCl2 on intracellular nucleotide concentration, extracellular nucleotide breakdown, mitochondrial function, and nitric oxide (NO) production in microvascular ECs. Moreover, we investigated how nucleotide precursor supplementation and adenosine deaminase inhibition protected against CoCl2-mediated disturbances. Mouse (H5V) and human (HMEC-1) microvascular ECs were exposed to CoCl2-mimicked hypoxia for 24 h in the presence of nucleotide precursors: adenine and ribose, and adenosine deaminase inhibitor, 2′deoxycoformycin. CoCl2 treatment decreased NO production by ECs, depleted intracellular ATP concentration, and increased extracellular nucleotide and adenosine catabolism in both H5V and HMEC-1 cell lines. Diminished intracellular ATP level was the effect of disturbed mitochondrial phosphorylation, while nucleotide precursors effectively restored the ATP pool via the salvage pathway and improved endothelial function under CoCl2 treatment. Endothelial protective effects of adenine and ribose were further enhanced by adenosine deaminase inhibition, that increased adenosine concentration. This work points to a novel strategy for protection of hypoxic ECs by replenishing the adenine nucleotide pool and promoting adenosine signaling.
Collapse
|
6
|
Salidroside attenuates CoCl 2-simulated hypoxia injury in PC12 cells partly by mitochondrial protection. Eur J Pharmacol 2021; 912:174617. [PMID: 34748770 DOI: 10.1016/j.ejphar.2021.174617] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023]
Abstract
Salidroside has been shown to exert neuroprotective effects against hypoxia. However, its mitochondrial protective mechanisms still remain elusive. The present study aimed to explore the mitochondrial protection of salidroside on PC12 cells and the involved mechanisms. The hypoxic injury of PC12 cells was triggered by CoCl2 stimulus. The contents of LDH release, SOD, GSH-PX, Na+-K+-ATPase, ATP, NAD+ and NADH were determined by using commercial biochemical kits. Clark-type oxygen electrode and Seahorse XFe24 analyzer were employed to evaluate cell respiration and measure oxygen consumption rate (OCR), respectively. Mitochondrial swelling and mitochondrial membrane potential (MMP) were measured by using isolated mitochondria from the brain tissue of mice. The proteins expression of cleaved Caspase-3, HIF-1α, ISCU1/2, COX10 and PFKP were tested by immunofluorescence and Western blot. While the genes expression of Caspase-3, HIF-1α, ISCU1/2, COX10 and miR-210 were tested by quantitative real-time PCR (qRT-PCR) analysis. Salidroside alleviated CoCl2-induced oxidative stress in PC12 cells as evidenced by increased cell viability, decreased LDH release and elevated GSH-PX and SOD activities. Salidroside could inhibit apoptosis by suppressing the level of cleaved Caspase-3 and Caspase-3. The enhanced mitochondrial energy synthesis by salidroside treatment was evidenced by the increases of Na+-K+-ATPase activity, ATP content, NAD+/NADH ratio, cellular respiration and OCR. In addition, salidroside could reduce mitochondrial swelling and MMP dissipation in isolated mitochondria. The results of immunofluorescence, Western blot and qRT-PCR analyses further revealed that salidroside raised the level of HIF-1α, ISCU1/2, COX10, and miR-210. Collectively, salidroside can reverse CoCl2-simulated hypoxia injury in PC12 cells partly by mitochondrial protection via inhibiting oxidative stress event, anti-apoptosis and enhancing mitochondrial energy synthesis.
Collapse
|
7
|
Strassheim D, Sullivan T, Irwin DC, Gerasimovskaya E, Lahm T, Klemm DJ, Dempsey EC, Stenmark KR, Karoor V. Metabolite G-Protein Coupled Receptors in Cardio-Metabolic Diseases. Cells 2021; 10:3347. [PMID: 34943862 PMCID: PMC8699532 DOI: 10.3390/cells10123347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have originally been described as a family of receptors activated by hormones, neurotransmitters, and other mediators. However, in recent years GPCRs have shown to bind endogenous metabolites, which serve functions other than as signaling mediators. These receptors respond to fatty acids, mono- and disaccharides, amino acids, or various intermediates and products of metabolism, including ketone bodies, lactate, succinate, or bile acids. Given that many of these metabolic processes are dysregulated under pathological conditions, including diabetes, dyslipidemia, and obesity, receptors of endogenous metabolites have also been recognized as potential drug targets to prevent and/or treat metabolic and cardiovascular diseases. This review describes G protein-coupled receptors activated by endogenous metabolites and summarizes their physiological, pathophysiological, and potential pharmacological roles.
Collapse
Affiliation(s)
- Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - David C. Irwin
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Tim Lahm
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Dwight J. Klemm
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Spatola BN, Lerner AG, Wong C, Dela Cruz T, Welch M, Fung W, Kovalenko M, Losenkova K, Yegutkin GG, Beers C, Corbin J, Soros VB. Fully human anti-CD39 antibody potently inhibits ATPase activity in cancer cells via uncompetitive allosteric mechanism. MAbs 2021; 12:1838036. [PMID: 33146056 PMCID: PMC7646477 DOI: 10.1080/19420862.2020.1838036] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extracellular ATP/adenosine axis in the tumor microenvironment (TME) has emerged as an important immune-regulatory pathway. Nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), otherwise known as CD39, is highly expressed in the TME, both on infiltrating immune cells and tumor cells across a broad set of cancer indications. CD39 processes pro-inflammatory extracellular ATP to ADP and AMP, which is then processed by Ecto-5ʹ-nucleotidase/CD73 to immunosuppressive adenosine. Directly inhibiting the enzymatic function of CD39 via an antibody has the potential to unleash an immune-mediated anti-tumor response via two mechanisms: 1) increasing the availability of immunostimulatory extracellular ATP released by damaged and/or dying cells, and 2) reducing the generation and accumulation of suppressive adenosine within the TME. Tizona Therapeutics has engineered a novel first-in-class fully human anti-CD39 antibody, TTX-030, that directly inhibits CD39 ATPase enzymatic function with sub-nanomolar potency. Further characterization of the mechanism of inhibition by TTX-030 using CD39+ human melanoma cell line SK-MEL-28 revealed an uncompetitive allosteric mechanism (α < 1). The uncompetitive mechanism of action enables TTX-030 to inhibit CD39 at the elevated ATP concentrations reported in the TME. Maximal inhibition of cellular CD39 ATPase velocity was 85%, which compares favorably to results reported for antibody inhibitors to other enzyme targets. The allosteric mechanism of TTX-030 was confirmed via mapping the epitope to a region of CD39 distant from its active site, which suggests possible models for how potent inhibition is achieved. In summary, TTX-030 is a potent allosteric inhibitor of CD39 ATPase activity that is currently being evaluated in clinical trials for cancer therapy.
Collapse
Affiliation(s)
- Bradley N Spatola
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Alana G Lerner
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - Clifford Wong
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Tracy Dela Cruz
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA.,Immunology, Trishula Therapeutics, South San Francisco , CA, USA
| | - Megan Welch
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - Wanchi Fung
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | | | | | | | - Courtney Beers
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - John Corbin
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Vanessa B Soros
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| |
Collapse
|
9
|
Hanidziar D, Robson SC. Synapomorphic features of hepatic and pulmonary vasculatures include comparable purinergic signaling responses in host defense and modulation of inflammation. Am J Physiol Gastrointest Liver Physiol 2021; 321:G200-G212. [PMID: 34105986 PMCID: PMC8410108 DOI: 10.1152/ajpgi.00406.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatosplanchnic and pulmonary vasculatures constitute synapomorphic, highly comparable networks integrated with the external environment. Given functionality related to obligatory requirements of "feeding and breathing," these organs are subject to constant environmental challenges entailing infectious risk, antigenic and xenobiotic exposures. Host responses to these stimuli need to be both protective and tightly regulated. These functions are facilitated by dualistic, high-low pressure blood supply of the liver and lungs, as well as tolerogenic characteristics of resident immune cells and signaling pathways. Dysregulation in hepatosplanchnic and pulmonary blood flow, immune responses, and microbiome implicate common pathogenic mechanisms across these vascular networks. Hepatosplanchnic diseases, such as cirrhosis and portal hypertension, often impact lungs and perturb pulmonary circulation and oxygenation. The reverse situation is also noted with lung disease resulting in hepatic dysfunction. Others, and we, have described common features of dysregulated cell signaling during liver and lung inflammation involving extracellular purines (e.g., ATP, ADP), either generated exogenously or endogenously. These metabokines serve as danger signals, when released by bacteria or during cellular stress and cause proinflammatory and prothrombotic signals in the gut/liver-lung vasculature. Dampening of these danger signals and organ protection largely depends upon activities of vascular and immune cell-expressed ectonucleotidases (CD39 and CD73), which convert ATP and ADP into anti-inflammatory adenosine. However, in many inflammatory disorders involving gut, liver, and lung, these protective mechanisms are compromised, causing perpetuation of tissue injury. We propose that interventions that specifically target aberrant purinergic signaling might prevent and/or ameliorate inflammatory disorders of the gut/liver and lung axis.
Collapse
Affiliation(s)
- Dusan Hanidziar
- 1Department of Anesthesia, Critical Care and Pain Medicine, grid.32224.35Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Simon C. Robson
- 2Department of Anesthesia, Critical Care and Pain Medicine, Center for Inflammation Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts,3Department of Medicine, Division of Gastroenterology/Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Toledo-Ibelles P, Gutiérrez-Vidal R, Calixto-Tlacomulco S, Delgado-Coello B, Mas-Oliva J. Hepatic Accumulation of Hypoxanthine: A Link Between Hyperuricemia and Nonalcoholic Fatty Liver Disease. Arch Med Res 2021; 52:692-702. [PMID: 33966916 DOI: 10.1016/j.arcmed.2021.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND An elevated level of plasma uric acid has been widely recognized as a risk factor for non-alcoholic fatty liver disease (NAFLD), where oxidative stress and inflammation play an important role in the pathophysiology of the disease. Although the complete molecular mechanisms involved remain unknown, while under physiological conditions uric acid presents antioxidant properties, hyperuricemia has been linked to oxidative stress, chronic low-grade inflammation, and insulin resistance, basic signs of NAFLD. AIM OF STUDY Employing in vivo experimentation, we aim to investigate whether a high-fat diet rich in cholesterol (HFD), modifies the metabolism of purines in close relationship to molecular events associated with the development of NAFLD. In vitro experiments employing HepG2 cells are also carried out to study the phenomenon of oxidative stress. METHODS Adult male rabbits were fed for 8 weeks an HFD to induce NAFLD. At the beginning of the experiment and every 15 d until the completion of the study, plasma levels of lipids, lipoproteins, and uric acid were measured. Liver tissue was isolated, and histology performed followed by the biochemical determination of hypoxanthine, protein expression of xanthine oxidoreductase (XOR) by western blot analysis, and xanthine oxidase (XO) activity using an enzymatic kinetic assay. Furthermore, we employed in vitro experimentation studying HepG2 cells to measure the effect of hypoxanthine and H2O2 upon the production of radical oxygen species (ROS), XO activity, and cell viability. RESULTS AND CONCLUSION Hepatic tissue from rabbits fed the HFD diet showed signs of NAFLD associated with an increased ROS concentration and an altered purine metabolism characterized by the increase in hypoxanthine, together with an apparent equilibrium displacement of XOR towards the xanthine dehydrogenase (XDH) isoform of the enzyme. This protein shift visualized by a western blot analysis, associated with an increase in plasma uric acid and hepatocyte hypoxanthine could be understood as a compensatory series of events secondary to the establishment of oxidative stress associated with the chronic establishment of fatty liver disease.
Collapse
Affiliation(s)
- Paola Toledo-Ibelles
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. Ciudad de México, México
| | - Roxana Gutiérrez-Vidal
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. Ciudad de México, México
| | - Sandra Calixto-Tlacomulco
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. Ciudad de México, México
| | - Blanca Delgado-Coello
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. Ciudad de México, México
| | - Jaime Mas-Oliva
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. Ciudad de México, México.
| |
Collapse
|
11
|
Caruso V, Zuccarini M, Di Iorio P, Muhammad I, Ronci M. Metabolic Changes Induced by Purinergic Signaling: Role in Food Intake. Front Pharmacol 2021; 12:655989. [PMID: 33995077 PMCID: PMC8117016 DOI: 10.3389/fphar.2021.655989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/09/2021] [Indexed: 01/30/2023] Open
Abstract
The purinergic signalling has a well-established role in the regulation of energy homeostasis, but there is growing evidence of its implication in the control of food intake. In this review, we provide an integrative view of the molecular mechanisms leading to changes in feeding behaviour within hypothalamic neurons following purinergic receptor activation. We also highlight the importance of purinergic signalling in metabolic homeostasis and the possibility of targeting its receptors for therapeutic purposes.
Collapse
Affiliation(s)
- Vanni Caruso
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia.,Institute for Research on Pain, ISAL-Foundation, Rimini, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy
| | - Ishaq Muhammad
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia
| | - Maurizio Ronci
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy.,Department of Pharmacy, University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
12
|
Yegutkin GG. Adenosine metabolism in the vascular system. Biochem Pharmacol 2020; 187:114373. [PMID: 33340515 DOI: 10.1016/j.bcp.2020.114373] [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: 11/04/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
The concept of extracellular purinergic signaling was first proposed by Geoffrey Burnstock in the early 1970s. Since then, extracellular ATP and its metabolites ADP and adenosine have attracted an enormous amount of attention in terms of their involvement in a wide range of immunomodulatory, thromboregulatory, angiogenic, vasoactive and other pathophysiological activities in different organs and tissues, including the vascular system. In addition to significant progress in understanding the properties of nucleotide- and adenosine-selective receptors, recent studies have begun to uncover the complexity of regulatory mechanisms governing the duration and magnitude of the purinergic signaling cascade. This knowledge has led to the development of new paradigms in understanding the entire purinome by taking into account the multitude of signaling and metabolic pathways involved in biological effects of ATP and adenosine and compartmentalization of the adenosine system. Along with the "canonical route" of ATP breakdown to adenosine via sequential ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities, it has now become clear that purine metabolism is the result of concerted effort between ATP release, its metabolism through redundant nucleotide-inactivating and counteracting ATP-regenerating ectoenzymatic pathways, as well as cellular nucleoside uptake and phosphorylation of adenosine to ATP through complex phosphotransfer reactions. In this review I provide an overview of key enzymes involved in adenosine metabolic network, with special emphasis on the emerging roles of purine-converting ectoenzymes as novel targets for cancer and vascular therapies.
Collapse
|
13
|
Casili G, Lanza M, Campolo M, Messina S, Scuderi S, Ardizzone A, Filippone A, Paterniti I, Cuzzocrea S, Esposito E. Therapeutic potential of flavonoids in the treatment of chronic venous insufficiency. Vascul Pharmacol 2020; 137:106825. [PMID: 33278582 DOI: 10.1016/j.vph.2020.106825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/24/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Chronic venous insufficiency (CVI) is a common disorder associated with a variety of symptoms in later disease stages; despite the high prevalence of this pathology, suitable pharmaceutical therapies have not been explored to date. In this context, it was recently reported that a chronic increase in venous wall stress or biomechanical stretch is sufficient to cause development of varicose veins. Recent evidence demonstrate that flavonoids are natural substances that convey the circulatory system functionality, playing a key role in blood flow. Particularly, troxerutin, diosmin and horse chestnut extract, appear protective for the management of vascular diseases. The aim of the present study was to evaluate the effect of a flavonoid compound, containing troxerutin, diosmin and horse chestnut extract on in vitro model on HUVECs cells, due to its production of vasculoregulatory and vasculotropic molecules, on an ex-vivo model on mesenteric vessel contraction, to regularize mesenteric microcirculation and on in vivo model of CVI-induced by saphene vein ligation. Furthermore, the flavonoid compound capacity of extensibility and compatibility with peripheral veins was investigated through a tissue block culture study. The degree of absorption, the contractile venous activity, the histological analysis, the immunoistochemical and immunofluorescence evaluation for VEGF and CD34 were performed, together with inflammatory mediators dosage. For the first time, this research revealed the therapeutic potential of a compound, enriched with flavonoids, to be a supportive treatment, suitable to reduce varicose vein pathophysiology and to regularize venous tone.
Collapse
Affiliation(s)
- Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Salvatore Messina
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Sarah Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy; Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO, USA
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy.
| |
Collapse
|
14
|
Losenkova K, Zuccarini M, Karikoski M, Laurila J, Boison D, Jalkanen S, Yegutkin GG. Compartmentalization of adenosine metabolism in cancer cells and its modulation during acute hypoxia. J Cell Sci 2020; 133:jcs241463. [PMID: 32317394 PMCID: PMC10681022 DOI: 10.1242/jcs.241463] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/02/2020] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine mediates diverse anti-inflammatory, angiogenic and vasoactive effects, and has become an important therapeutic target for cancer, which has been translated into clinical trials. This study was designed to comprehensively assess adenosine metabolism in prostate and breast cancer cells. We identified cellular adenosine turnover as a complex cascade, comprising (1) the ectoenzymatic breakdown of ATP via sequential ecto-nucleotide pyrophosphatase/phosphodiesterase-1 (NPP1, officially known as ENPP1), ecto-5'-nucleotidase (CD73, also known as NT5E), and adenosine deaminase reactions, and ATP re-synthesis through a counteracting adenylate kinase and members of the nucleoside diphosphate kinase (NDPK, also known as NME/NM23) family; (2) the uptake of nucleotide-derived adenosine via equilibrative nucleoside transporters; and (3) the intracellular adenosine phosphorylation into ATP by adenosine kinase and other nucleotide kinases. The exposure of cancer cells to 1% O2 for 24 h triggered an ∼2-fold upregulation of CD73, without affecting nucleoside transporters, adenosine kinase activity and cellular ATP content. The ability of adenosine to inhibit the tumor-initiating potential of breast cancer cells via a receptor-independent mechanism was confirmed in vivo using a xenograft mouse model. The existence of redundant pathways controlling extracellular and intracellular adenosine provides a sufficient justification for reexamination of the current concepts of cellular purine homeostasis and signaling in cancer.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Mariachiara Zuccarini
- MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
- Department of Medical, Oral and Biotechnological Sciences, 'G. D'Annunzio' University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marika Karikoski
- MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Juha Laurila
- MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson and New Jersey Medical Schools, Rutgers University, Piscataway, NJ 08854, USA
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | | |
Collapse
|
15
|
Shen Y, Zhai P, Xi P, Chen C, Mou F, Li F, Zhang J. Identification of new trace related impurities in adenosine products using ultra-high-performance liquid chromatography/electrospray ionization quadrupole time-of-flight tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2019; 33:1739-1750. [PMID: 31146299 DOI: 10.1002/rcm.8487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 06/09/2023]
Abstract
RATIONALE Adenosine can induce various physiopathological effects and has been adopted as a drug to treat certain forms of supraventricular tachycardia. Adenosine is predominantly produced via fermentation that may generate many other bioactive compounds with similar structures at trace levels. Thus, it is necessary to identify these trace structurally related impurities. A new method using ultra-high-performance liquid chromatography (UHPLC) coupled with quadrupole time-of-flight tandem mass spectrometry (QTOF-MS/MS) is proposed to separate complex components at trace amounts; to obtain accurate mass measurements providing the elemental composition of unknown compounds; and MS/MS spectra for elucidation of structures. METHODS A UHPLC/QTOF-MS method was developed to separate and detect trace related impurities with structures similar to adenosine in formation samples. MS and MS/MS spectra of the impurities detected in real samples were acquired and used to propose their structures. Available reference standards were used to confirm the identification of some of the impurities detected. RESULTS Nine trace impurities of adenosine were separated and characterized. Of these nine, five were confirmed as 5'-adenylic acid, hypoxanthine, inosine, adenine, and 2'-deoxyadenosine by comparison with their available reference standards. The remaining four were proposed to be ADP-ribose, two S-epimers of 5'-deoxy-5'-methylsulfinyl adenosine, and 3'-α-glucosyl adenosine based on their MS and MS/MS spectra, and available literature. Additionally, the MS/MS fingerprints of the monosaccharide glycosyl groups were discovered and discussed. CONCLUSIONS A UHPLC/QTOF-MS method was established and used for the separation and characterization of nine trace related impurities of adenosine. Their structures were identified based on the MS and MS/MS spectra and retention times. In addition, the specific MS/MS fingerprints of the monosaccharide glycosyl moieties of the adenosine impurity analogs were summarized.
Collapse
Affiliation(s)
- Yeming Shen
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| | - Peilu Zhai
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| | - Pengxuan Xi
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| | - Chen Chen
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| | - Fangjing Mou
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| | - Fuxin Li
- Jiyuan Branch, Henan Tobacco Corporation, 38 Huanghe Road, 459000, Jiyuan, China
| | - Jianye Zhang
- College of Chemistry and Molecular Engineering, Zhengzhou University, 100 Science Road, 450001, Zhengzhou, China
| |
Collapse
|
16
|
You S, Qian J, Wu G, Qian Y, Wang Z, Chen T, Wang J, Huang W, Liang G. Schizandrin B attenuates angiotensin II induced endothelial to mesenchymal transition in vascular endothelium by suppressing NF-κB activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152955. [PMID: 31146168 DOI: 10.1016/j.phymed.2019.152955] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Angiotensin II (Ang II)-induced chronic inflammation and oxidative stress often leads to irreversible vascular injury, in which the endothelial to mesenchymal transition (EndMT) in the endothelial layers are involved. Schisandrin B (Sch B), a natural product isolated from traditional Schisandra chinensis, has been reported to exert vascular protective properties with unclear mechanism. HYPOTHESIS/PURPOSE This study investigated the protective effects and mechanism of Sch B against Ang II-induced vascular injury. METHODS C57BL/6 mice were subcutaneous injected of Ang II for 4 weeks to induce irreversible vascular injury. In vitro, Ang II-induced HUVECs injury was used to study the underlying mechanism. The markers of EndMT, inflammation and oxidative stress were studied both in vitro and in vivo. RESULTS Pre-administration of Sch B effectively attenuated phenotypes of vascular EndMT and fibrosis in Ang II-treated animals, accompanied with decreased inflammatory cytokine and ROS. The in vitro data from HUVECs suggest that Sch B directly targets NF-κB activation to suppress Ang II-induced EndMT and vascular injury. The activation of EndMT in the presence of Ang II is regulated by the NF-κB, a common player in inflammation and oxidative stress. Ang II-induced inflammation and oxidative stress also contributed to vascular EndMT development and Sch B inhibited inflammation/ROS-mediated EndMT by suppressing NF-κB. CONCLUSION EndMT contributes to vascular injury in Ang II-treated mice, and it can be prevented via suppressing NF-κB activation by Sch B treatment. These results also imply that NF-κB might be a promising target to attenuate vascular remodeling induced by inflammation and oxidative stress through an EndMT mechanism.
Collapse
Affiliation(s)
- Shengban You
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanyuan Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengxian Wang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Taiwei Chen
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingying Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|