1
|
Verma DK, Chaudhary S, Sunil S. Investigation of endocytic pathways during entry of RNA viruses reveal novel host proteins as lipid raft dependent endocytosis mediators. Virology 2025; 608:110531. [PMID: 40262431 DOI: 10.1016/j.virol.2025.110531] [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: 11/12/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Abstract
Entry of viruses inside host cell after successful attachment is an essential step to ensure its genome replication and progeny production using host cell machinery. Targeting viral entry has been proven an effective therapeutic approach to prevent or treat viral infections. Viruses exploit different operational ligand entry routes to gain entry inside the host cell. Host membrane rafts are crucial for membrane mediated events such as ligand binding and internalization, signaling and pathogen entry. However, those host proteins involved in this phenomenon and molecular mechanism of this mode of endocytosis has not yet been elucidated. In present study, we investigated raft-dependent endocytosis as a major route for host cell entry for three different enveloped viruses viz. SARS-CoV-2, DENV and CHIKV. Subsequently, we performed quantitative global proteomics of SARS-CoV-2 infected Vero cells at the time of virus entry and during peak viral infection and compared proteomic changes with uninfected control. Subsequently, we implemented pathway enrichment of differentially regulated host proteins and identified regulated cellular pathways during different stages of infection. Finally, we investigated the role of selected proteins identified as significantly regulated through proteome analysis along with some of those proteins previously reported to be involved in any mode of endocytosis, in the raft-dependent endocytosis using inhibitor assay and further validated their role in viral entry through loss-of-function assays. Our results confirm that enveloped viruses exploit the raft-dependent endocytosis as a major route for host cell entry. We further report novel host cell proteins that participate as mediators of raft-dependent endocytosis.
Collapse
Affiliation(s)
- Dileep Kumar Verma
- Vector-Borne Disease Group, International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sakshi Chaudhary
- Vector-Borne Disease Group, International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sujatha Sunil
- Vector-Borne Disease Group, International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India.
| |
Collapse
|
2
|
Reed EC, Kim JD, Case AJ. Non-canonical hemoglobin: An updated review on its ubiquitous expression. Redox Biol 2025; 82:103602. [PMID: 40138914 PMCID: PMC11984994 DOI: 10.1016/j.redox.2025.103602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Hemoglobin, once thought to be exclusive to erythrocytes, has been identified to be expressed in various cell types over the past several decades. While hemoglobin's function within erythrocytes is primarily characterized as a gaseous transport molecule, its function within non-erythrocyte cells varies among different cell types, and in many cases, remains to be fully elucidated. Despite this variability, hemoglobin expression seems to broadly function as a redox modulator, whether it is involved in the hypoxic response, mitochondrial function, antioxidant balance or, like in erythrocytes, gas transport. This review provides an updated summary of the most recent discoveries of hemoglobin in non-erythrocyte cells. While discussing the function and regulation of this ubiquitous protein, we additionally compare these cell-specific details to identify commonalities throughout the diverse group of hemoglobin-expressing cells. Lastly, we discuss potential implications of non-canonical hemoglobin in various disease states such neurodegeneration, autoimmune disorders, psychological trauma, and hemoglobinopathies, while providing future directions for hemoglobin research.
Collapse
Affiliation(s)
- Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | | | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
3
|
Reed EC, Silva VA, Giebel KR, Natour T, Lauten TH, Jojo CN, Schlieker AE, Case AJ. Hemoglobin alpha is a redox-sensitive mitochondrial-related protein in T-lymphocytes. Free Radic Biol Med 2025; 227:1-11. [PMID: 39586383 PMCID: PMC11757050 DOI: 10.1016/j.freeradbiomed.2024.11.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/08/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Hemoglobin subunits, which form the well-characterized, tetrameric, oxygen-carrying protein, have recently been described to be expressed in various non-canonical cell types. However, the exact function of hemoglobin subunits within these cells remains to be fully elucidated. Herein, we report for the first time, the expression of hemoglobin alpha-a1 (Hba-a1) in T-lymphocytes and describe its role as a mitochondrial-associated antioxidant. Within naïve T-lymphocytes, Hba-a1 mRNA and HBA protein are present and highly induced by redox perturbations, particularly those arising from the mitochondria. Additionally, preliminary data using a T-lymphocyte specific Hba-a1 knock-out mouse model indicated that the loss of Hba-a1 led to an exacerbated production of mitochondrial reactive oxygen species and inflammatory cytokines after a stress challenge, further supporting the role of HBA acting to buffer the mitochondrial redox environment. Interestingly, we observed Hba-a1 expression to be significantly upregulated or downregulated depending on T-lymphocyte polarization and metabolic state, which appeared to be controlled by both transcriptional regulation and chromatin remodeling. Altogether, these data suggest Hba-a1 may function as a crucial mitochondrial-associated antioxidant and appears to possess critical and complex functions related to T-lymphocyte activation and differentiation.
Collapse
Affiliation(s)
- Emily C Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Valeria A Silva
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Kristen R Giebel
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Tatlock H Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Caroline N Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Abigail E Schlieker
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - Adam J Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, USA; Department of Medical Physiology, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
4
|
Lang A, Collins JM, Nijsure MP, Belali S, Khan MP, Moharrer Y, Schipani E, Yien YY, Fan Y, Gelinsky M, Vinogradov SA, Koch C, Boerckel JD. Local erythropoiesis directs oxygen availability in bone fracture repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632440. [PMID: 39829797 PMCID: PMC11741344 DOI: 10.1101/2025.01.10.632440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Bone fracture ruptures blood vessels and disrupts the bone marrow, the site of new red blood cell production (erythropoiesis). Current dogma holds that bone fracture causes severe hypoxia at the fracture site, due to vascular rupture, and that this hypoxia must be overcome for regeneration. Here, we show that the early fracture site is not hypoxic, but instead exhibits high oxygen tension (> 55 mmHg, or 8%), similar to the red blood cell reservoir, the spleen. This elevated oxygen stems not from angiogenesis but from activated erythropoiesis in the adjacent bone marrow. Fracture-activated erythroid progenitor cells concentrate oxygen through haemoglobin formation. Blocking transferrin receptor 1 (CD71)-mediated iron uptake prevents oxygen binding by these cells, induces fracture site hypoxia, and enhances bone repair through increased angiogenesis and osteogenesis. These findings upend our current understanding of the early phase of bone fracture repair, provide a mechanism for high oxygen tension in the bone marrow after injury, and reveal an unexpected and targetable role of erythroid progenitors in fracture repair.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Joseph M. Collins
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P. Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Simin Belali
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Yvette Y. Yien
- Division of Hematology/Oncology, Department of Medicine and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sergei A. Vinogradov
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Cameron Koch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Reed EC, Silva VA, Giebel KR, Natour T, Lauten TH, Jojo CN, Schleiker AE, Case AJ. Hemoglobin alpha is a redox-sensitive mitochondrial-related protein in T-lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613298. [PMID: 39345360 PMCID: PMC11429782 DOI: 10.1101/2024.09.16.613298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hemoglobin subunits, which form the well-characterized, tetrameric, oxygen-carrying protein, have recently been described to be expressed in various non-canonical cell types. However, the exact function of hemoglobin subunits within these cells remains to be fully elucidated. Herein, we report for the first time, the expression of hemoglobin alpha-a1 (Hba-a1) in T-lymphocytes and describe its role as a mitochondrial-associated antioxidant. Within naïve T-lymphocytes, Hba-a1 mRNA and HBA protein are present and highly induced by redox perturbations, particularly those arising from the mitochondria. Additionally, preliminary data using a T-lymphocyte specific Hba-a1 knock-out mouse model indicated that the loss of Hba-a1 led to an exacerbated production of mitochondrial reactive oxygen species and inflammatory cytokines after a stress challenge, further supporting the role of HBA acting to buffer the mitochondrial redox environment. Interestingly, we observed Hba-a1 expression to be significantly upregulated or downregulated depending on T-lymphocyte polarization and metabolic state, which appeared to be controlled by both transcriptional regulation and chromatin remodeling. Altogether, these data suggest Hba-a1 may function as a crucial mitochondrial-associated antioxidant and appears to possess critical and complex functions related to T-lymphocyte activation and differentiation.
Collapse
Affiliation(s)
- Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Valeria A. Silva
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Kristen R. Giebel
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Caroline N. Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Abigail E. Schleiker
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
6
|
Abbineni PS, Baid S, Weiss MJ. A moonlighting job for α-globin in blood vessels. Blood 2024; 144:834-844. [PMID: 38848504 PMCID: PMC11830976 DOI: 10.1182/blood.2023022192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
ABSTRACT Red blood cells express high levels of hemoglobin A tetramer (α2β2) to facilitate oxygen transport. Hemoglobin subunits and related proteins are also expressed at lower levels in other tissues across the animal kingdom. Physiological functions for most nonerythroid globins likely derive from their ability to catalyze reduction-oxidation (redox) reactions via electron transfer through heme-associated iron. An interesting example is illustrated by the recent discovery that α-globin without β-globin is expressed in some arteriolar endothelial cells (ECs). α-globin binds EC nitric oxide (NO) synthase (eNOS) and degrades its enzymatic product NO, a potent vasodilator. Thus, depletion of α-globin in ECs or inhibition of its association with eNOS causes arteriolar relaxation and lowering of blood pressure in mice. Some of these findings have been replicated in isolated human blood vessels, and genetic studies are tractable in populations in which α-thalassemia alleles are prevalent. Two small studies identified associations between loss of α-globin genes in humans and NO-regulated vascular responses elicited by local hypoxia-induced blood flow or thermal stimulation. In a few larger population-based studies, no associations were detected between loss of α-globin genes and blood pressure, ischemic stroke, or pulmonary hypertension. In contrast, a significant positive association between α-globin gene copy number and kidney disease was detected in an African American cohort. Further studies are required to define comprehensively the expression of α-globin in different vascular beds and ascertain their overall impact on normal and pathological vascular physiology.
Collapse
Affiliation(s)
- Prabhodh S. Abbineni
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Srishti Baid
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Mitchell J. Weiss
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
7
|
Eltobgy M, Johns F, Farkas D, Leuenberger L, Cohen SP, Ho K, Karow S, Swoope G, Pannu S, Horowitz JC, Mallampalli RK, Englert JA, Bednash JS. Longitudinal transcriptomic analysis reveals persistent enrichment of iron homeostasis and erythrocyte function pathways in severe COVID-19 ARDS. Front Immunol 2024; 15:1397629. [PMID: 39161760 PMCID: PMC11330807 DOI: 10.3389/fimmu.2024.1397629] [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: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Introduction The acute respiratory distress syndrome (ARDS) is a common complication of severe COVID-19 and contributes to patient morbidity and mortality. ARDS is a heterogeneous syndrome caused by various insults, and results in acute hypoxemic respiratory failure. Patients with ARDS from COVID-19 may represent a subgroup of ARDS patients with distinct molecular profiles that drive disease outcomes. Here, we hypothesized that longitudinal transcriptomic analysis may identify distinct dynamic pathobiological pathways during COVID-19 ARDS. Methods We identified a patient cohort from an existing ICU biorepository and established three groups for comparison: 1) patients with COVID-19 ARDS that survived hospitalization (COVID survivors, n = 4), 2) patients with COVID-19 ARDS that did not survive hospitalization (COVID non-survivors, n = 5), and 3) patients with ARDS from other causes as a control group (ARDS controls, n = 4). RNA was isolated from peripheral blood mononuclear cells (PBMCs) at 4 time points (Days 1, 3, 7, and 10 following ICU admission) and analyzed by bulk RNA sequencing. Results We first compared transcriptomes between groups at individual timepoints and observed significant heterogeneity in differentially expressed genes (DEGs). Next, we utilized the likelihood ratio test to identify genes that exhibit different patterns of change over time between the 3 groups and identified 341 DEGs across time, including hemoglobin subunit alpha 2 (HBA1, HBA2), hemoglobin subunit beta (HBB), von Willebrand factor C and EGF domains (VWCE), and carbonic anhydrase 1 (CA1), which all demonstrated persistent upregulation in the COVID non-survivors compared to COVID survivors. Of the 341 DEGs, 314 demonstrated a similar pattern of persistent increased gene expression in COVID non-survivors compared to survivors, associated with canonical pathways of iron homeostasis signaling, erythrocyte interaction with oxygen and carbon dioxide, erythropoietin signaling, heme biosynthesis, metabolism of porphyrins, and iron uptake and transport. Discussion These findings describe significant differences in gene regulation during patient ICU course between survivors and non-survivors of COVID-19 ARDS. We identified multiple pathways that suggest heme and red blood cell metabolism contribute to disease outcomes. This approach is generalizable to larger cohorts and supports an approach of longitudinal sampling in ARDS molecular profiling studies, which may identify novel targetable pathways of injury and resolution.
Collapse
Affiliation(s)
- Moemen Eltobgy
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Finny Johns
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Daniela Farkas
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Laura Leuenberger
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Sarah P. Cohen
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Kevin Ho
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Sarah Karow
- Clinical Trials Management Office, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Gabrielle Swoope
- Clinical Trials Management Office, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Sonal Pannu
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Jeffrey C. Horowitz
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Rama K. Mallampalli
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Joshua A. Englert
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Joseph S. Bednash
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- The Center for RNA Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
8
|
Ma SY, Liu YT, Cun YS, Wang Q, Fu MC, Wu KD, Cai XY, Cheng ST, Patel N, Da M, Hu L, Deqin Z, Kang XJ, Yang M, Mo XM. Preoperative serum cortisone levels are associated with cognition in preschool-aged children with tetralogy of Fallot after corrective surgery: new evidence from human populations and mice. World J Pediatr 2024; 20:173-184. [PMID: 37737505 PMCID: PMC10884142 DOI: 10.1007/s12519-023-00754-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 08/06/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease. Children with TOF would be confronted with neurological impairment across their lifetime. Our study aimed to identify the risk factors for cerebral morphology changes and cognition in postoperative preschool-aged children with TOF. METHODS We used mass spectrometry (MS) technology to assess the levels of serum metabolites, Wechsler preschool and primary scale of intelligence-Fourth edition (WPPSI-IV) index scores to evaluate neurodevelopmental levels and multimodal magnetic resonance imaging (MRI) to detect cortical morphological changes. RESULTS Multiple linear regression showed that preoperative levels of serum cortisone were positively correlated with the gyrification index of the left inferior parietal gyrus in children with TOF and negatively related to their lower visual spaces index and nonverbal index. Meanwhile, preoperative SpO2 was negatively correlated with levels of serum cortisone after adjusting for all covariates. Furthermore, after intervening levels of cortisone in chronic hypoxic model mice, total brain volumes were reduced at both postnatal (P) 11.5 and P30 days. CONCLUSIONS Our results suggest that preoperative serum cortisone levels could be used as a biomarker of neurodevelopmental impairment in children with TOF. Our study findings emphasized that preoperative levels of cortisone could influence cerebral development and cognition abilities in children with TOF.
Collapse
Affiliation(s)
- Si-Yu Ma
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yu-Ting Liu
- Department of Radiology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yue-Shuang Cun
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Ming-Cui Fu
- Department of Radiology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Ke-De Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xin-Yu Cai
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Shu-Ting Cheng
- Department of Radiology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Min Da
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Liang Hu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Zhuoga Deqin
- Medical School of Nanjing University, Nanjing, 210093, China
| | - Xue-Jun Kang
- Key Laboratory of Child Development and Learning Science, Research Center For Learning Science, School of Biological Sciences & Medical Engineering, Ministry of Education, Southeast University, Nanjing, 210096, China.
| | - Ming Yang
- Department of Radiology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| | - Xu-Ming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
- Medical School of Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
9
|
Zhang F, Zhang B, Wang Y, Jiang R, Liu J, Wei Y, Gao X, Zhu Y, Wang X, Sun M, Kang J, Liu Y, You G, Wei D, Xin J, Bao J, Wang M, Gu Y, Wang Z, Ye J, Guo S, Huang H, Sun Q. An extra-erythrocyte role of haemoglobin body in chondrocyte hypoxia adaption. Nature 2023; 622:834-841. [PMID: 37794190 PMCID: PMC10600011 DOI: 10.1038/s41586-023-06611-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
Although haemoglobin is a known carrier of oxygen in erythrocytes that functions to transport oxygen over a long range, its physiological roles outside erythrocytes are largely elusive1,2. Here we found that chondrocytes produced massive amounts of haemoglobin to form eosin-positive bodies in their cytoplasm. The haemoglobin body (Hedy) is a membraneless condensate characterized by phase separation. Production of haemoglobin in chondrocytes is controlled by hypoxia and is dependent on KLF1 rather than the HIF1/2α pathway. Deletion of haemoglobin in chondrocytes leads to Hedy loss along with severe hypoxia, enhanced glycolysis and extensive cell death in the centre of cartilaginous tissue, which is attributed to the loss of the Hedy-controlled oxygen supply under hypoxic conditions. These results demonstrate an extra-erythrocyte role of haemoglobin in chondrocytes, and uncover a heretofore unrecognized mechanism in which chondrocytes survive a hypoxic environment through Hedy.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China.
| | - Bo Zhang
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing, China
- Nanhu Laboratory, Jiaxing, China
| | - Yuying Wang
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Runmin Jiang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jin Liu
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Yuexian Wei
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
- Nanhu Laboratory, Jiaxing, China
| | - Xinyue Gao
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
- Nanhu Laboratory, Jiaxing, China
| | - Yichao Zhu
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
- Nanhu Laboratory, Jiaxing, China
| | - Xinli Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mao Sun
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
| | - Junjun Kang
- Department of Neurobiology, The Fourth Military Medical University, Xi'an, China
| | - Yingying Liu
- Department of Neurobiology, The Fourth Military Medical University, Xi'an, China
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Ding Wei
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, China
| | - Jiajia Xin
- Department of Blood Transfusion, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Junxiang Bao
- Department of Aerospace Hygiene, The Fourth Military Medical University, Xi'an, China
| | - Meiqing Wang
- Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yu Gu
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Zhe Wang
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Jing Ye
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Shuangping Guo
- Department of Pathology, School of Basic Medicine and Xijing Hospital, State Key Laboratory of Cancer Biology, Air Force Medical Center, The Fourth Military Medical University, Xi'an, China
| | - Hongyan Huang
- Department of Oncology, Beijing Shijitan Hospital of Capital Medical University, Beijing, China
| | - Qiang Sun
- Frontier Biotechnology Laboratory, Beijing Institute of Biotechnology, Academy of Military Medical Science; Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China.
- Nanhu Laboratory, Jiaxing, China.
| |
Collapse
|
10
|
Rochín-Hernández LJ, Jiménez-Acosta MA, Ramírez-Reyes L, Figueroa-Corona MDP, Sánchez-González VJ, Orozco-Barajas M, Meraz-Ríos MA. The Proteome Profile of Olfactory Ecto-Mesenchymal Stem Cells-Derived from Patients with Familial Alzheimer's Disease Reveals New Insights for AD Study. Int J Mol Sci 2023; 24:12606. [PMID: 37628788 PMCID: PMC10454072 DOI: 10.3390/ijms241612606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disease and the first cause of dementia worldwide, has no effective treatment, and its pathological mechanisms are not yet fully understood. We conducted this study to explore the proteomic differences associated with Familial Alzheimer's Disease (FAD) in olfactory ecto-mesenchymal stem cells (MSCs) derived from PSEN1 (A431E) mutation carriers compared with healthy donors paired by age and gender through two label-free liquid chromatography-mass spectrometry approaches. The first analysis compared carrier 1 (patient with symptoms, P1) and its control (healthy donor, C1), and the second compared carrier 2 (patient with pre-symptoms, P2) with its respective control cells (C2) to evaluate whether the protein alterations presented in the symptomatic carrier were also present in the pre-symptom stages. Finally, we analyzed the differentially expressed proteins (DEPs) for biological and functional enrichment. These proteins showed impaired expression in a stage-dependent manner and are involved in energy metabolism, vesicle transport, actin cytoskeleton, cell proliferation, and proteostasis pathways, in line with previous AD reports. Our study is the first to conduct a proteomic analysis of MSCs from the Jalisco FAD patients in two stages of the disease (symptomatic and presymptomatic), showing these cells as a new and excellent in vitro model for future AD studies.
Collapse
Affiliation(s)
- Lory J. Rochín-Hernández
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Miguel A. Jiménez-Acosta
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Lorena Ramírez-Reyes
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Ciudad de México 07360, Mexico;
| | - María del Pilar Figueroa-Corona
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| | - Víctor J. Sánchez-González
- Centro Universitario de Los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Mexico; (V.J.S.-G.); (M.O.-B.)
| | - Maribel Orozco-Barajas
- Centro Universitario de Los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Mexico; (V.J.S.-G.); (M.O.-B.)
| | - Marco A. Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico; (L.J.R.-H.); (M.A.J.-A.); (M.d.P.F.-C.)
| |
Collapse
|
11
|
Wei KH, Lin IT, Chowdhury K, Lim KL, Liu KT, Ko TM, Chang YM, Yang KC, Lai SL(B. Comparative single-cell profiling reveals distinct cardiac resident macrophages essential for zebrafish heart regeneration. eLife 2023; 12:e84679. [PMID: 37498060 PMCID: PMC10411971 DOI: 10.7554/elife.84679] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/26/2023] [Indexed: 07/28/2023] Open
Abstract
Zebrafish exhibit a robust ability to regenerate their hearts following injury, and the immune system plays a key role in this process. We previously showed that delaying macrophage recruitment by clodronate liposome (-1d_CL, macrophage-delayed model) impairs neutrophil resolution and heart regeneration, even when the infiltrating macrophage number was restored within the first week post injury (Lai et al., 2017). It is thus intriguing to learn the regenerative macrophage property by comparing these late macrophages vs. control macrophages during cardiac repair. Here, we further investigate the mechanistic insights of heart regeneration by comparing the non-regenerative macrophage-delayed model with regenerative controls. Temporal RNAseq analyses revealed that -1d_CL treatment led to disrupted inflammatory resolution, reactive oxygen species homeostasis, and energy metabolism during cardiac repair. Comparative single-cell RNAseq profiling of inflammatory cells from regenerative vs. non-regenerative hearts further identified heterogeneous macrophages and neutrophils, showing alternative activation and cellular crosstalk leading to neutrophil retention and chronic inflammation. Among macrophages, two residential subpopulations (hbaa+ Mac and timp4.3+ Mac 3) were enriched only in regenerative hearts and barely recovered after +1d_CL treatment. To deplete the resident macrophage without delaying the circulating macrophage recruitment, we established the resident macrophage-deficient model by administrating CL earlier at 8 d (-8d_CL) before cryoinjury. Strikingly, resident macrophage-deficient zebrafish still exhibited defects in revascularization, cardiomyocyte survival, debris clearance, and extracellular matrix remodeling/scar resolution without functional compensation from the circulating/monocyte-derived macrophages. Our results characterized the diverse function and interaction between inflammatory cells and identified unique resident macrophages prerequisite for zebrafish heart regeneration.
Collapse
Affiliation(s)
- Ke-Hsuan Wei
- Graduate Institute of Life Sciences, National Defense Medical CenterTaipeiTaiwan
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - I-Ting Lin
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Kaushik Chowdhury
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Khai Lone Lim
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Kuan-Ting Liu
- Department of Biological Science & Technology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tai-Ming Ko
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Department of Biological Science & Technology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
| | - Kai-Chien Yang
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of MedicineTaipeiTaiwan
| | - Shih-Lei (Ben) Lai
- Graduate Institute of Life Sciences, National Defense Medical CenterTaipeiTaiwan
- Institute of Biomedical Sciences, Academia SinicaTaipeiTaiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
12
|
Li J, Ruggiero-Ruff RE, He Y, Qiu X, Lainez N, Villa P, Godzik A, Coss D, Nair MG. Sexual dimorphism in obesity is governed by RELMα regulation of adipose macrophages and eosinophils. eLife 2023; 12:e86001. [PMID: 37162190 PMCID: PMC10171862 DOI: 10.7554/elife.86001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/18/2023] [Indexed: 05/11/2023] Open
Abstract
Obesity incidence is increasing worldwide with the urgent need to identify new therapeutics. Sex differences in immune cell activation drive obesity-mediated pathologies where males are more susceptible to obesity comorbidities and exacerbated inflammation. Here, we demonstrate that the macrophage-secreted protein RELMα critically protects females against high-fat diet (HFD)-induced obesity. Compared to male mice, serum RELMα levels were higher in both control and HFD-fed females and correlated with frequency of adipose macrophages and eosinophils. RELMα-deficient females gained more weight and had proinflammatory macrophage accumulation and eosinophil loss in the adipose stromal vascular fraction (SVF), while RELMα treatment or eosinophil transfer rescued this phenotype. Single-cell RNA-sequencing of the adipose SVF was performed and identified sex and RELMα-dependent changes. Genes involved in oxygen sensing and iron homeostasis, including hemoglobin and lncRNA Gm47283/Gm21887, correlated with increased obesity, while eosinophil chemotaxis and response to amyloid-beta were protective. Monocyte-to-macrophage transition was also dysregulated in RELMα-deficient animals. Collectively, these studies implicate a RELMα-macrophage-eosinophil axis in sex-specific protection against obesity and uncover new therapeutic targets for obesity.
Collapse
Affiliation(s)
- Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Yuxin He
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Xinru Qiu
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California RiversideRiversideUnited States
| | - Nancy Lainez
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Pedro Villa
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California RiversideRiversideUnited States
| |
Collapse
|
13
|
Hansakon A, Ngamphiw C, Tongsima S, Angkasekwinai P. Arginase 1 Expression by Macrophages Promotes Cryptococcus neoformans Proliferation and Invasion into Brain Microvascular Endothelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:408-419. [PMID: 36548474 DOI: 10.4049/jimmunol.2200592] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Cryptococcal meningoencephalitis caused by Cryptococcus neoformans infection is the most common cause of death in HIV/AIDS patients. Macrophages are pivotal for the regulation of immune responses to cryptococcal infection by either playing protective function or facilitating fungal dissemination. However, the mechanisms underlying macrophage responses to C. neoformans remain unclear. To analyze the transcriptomic changes and identify the pathogenic factors of macrophages, we performed a comparative transcriptomic analysis of alveolar macrophage responses during C. neoformans infection. Alveolar macrophages isolated from C. neoformans-infected mice showed dynamic gene expression patterns, with expression change from a protective M1 (classically activated)-like to a pathogenic M2 (alternatively activated)-like phenotype. Arg1, the gene encoding the enzyme arginase 1, was found as the most upregulated gene in alveolar macrophages during the chronic infection phase. The in vitro inhibition of arginase activity resulted in a reduction of cryptococcal phagocytosis, intracellular growth, and proliferation, coupled with an altered macrophage response from pathogenic M2 to a protective M1 phenotype. In an in vitro model of the blood-brain barrier, macrophage-derived arginase was found to be required for C. neoformans invasion of brain microvascular endothelium. Further analysis of the degree of virulence indicated a positive correlation between arginase 1 expression in macrophages and cryptococcal brain dissemination in vivo. Thus, our data suggest that a dynamic macrophage activation that involves arginase expression may contribute to the cryptococcal disease by promoting cryptococcal growth, proliferation, and the invasion to the brain endothelium.
Collapse
Affiliation(s)
- Adithap Hansakon
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand.,Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Chumpol Ngamphiw
- National Biobank of Thailand, National Science and Technology Development Agency, Pathum Thani, Thailand; and
| | - Sissades Tongsima
- National Biobank of Thailand, National Science and Technology Development Agency, Pathum Thani, Thailand; and
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand.,Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
14
|
Chu M, Gao H, Esparza P, Pajulas A, Wang J, Kharwadkar R, Gao H, Kaplan MH, Tepper RS. Chronic developmental hypoxia alters rat lung immune cell transcriptomes during allergic airway inflammation. Physiol Rep 2023; 11:e15600. [PMID: 36750205 PMCID: PMC9904961 DOI: 10.14814/phy2.15600] [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: 11/28/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023] Open
Abstract
Populations that are born and raised at high altitude develop under conditions of chronic developmental hypoxia (CDH), which results in pulmonary adaptations of increased lung volume and diffusion capacity to increase gas exchange. It is not clear how CDH may alter allergic inflammation in the lung. In this study, we sought to characterize the impact of CDH on immune cell populations in the rat lung during a murine model of asthma. Rats were bred and raised in either hypoxic (15% oxygen, CDH) or normobaric room air (20% oxygen). At 3-weeks of age, animals were sensitized to ovalbumin (OVA) or physiologic saline (phosphate-buffered saline [PBS]) as a control, followed by three consecutive days of intra-nasal OVA or PBS at 6-weeks of age. We then assessed airway reactivity and allergic-associated cytokine levels. This was followed by single-cell transcriptomic profiling of lung cell populations. In scRNA-seq analysis, we assessed differentially expressed genes, differentially enriched functional pathways, immune cell exhaustion/activation markers, and immune cell secretory products. Our results show that while OVA heightened airway reactivity, CDH suppressed airway reactivity in OVA-challenged and control animals. Through scRNA-seq analysis, we further demonstrate that CDH alters the transcriptional landscape in the lung and alters transcriptional programs in immune cells. These data define CDH-dependent changes in the lung that impact airway reactivity.
Collapse
Affiliation(s)
- Michelle Chu
- Department of Microbiology and ImmunologyIndiana UniversityIndianapolisIndianaUSA
| | - Huanling Gao
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana UniversityIndianapolisIndianaUSA
| | - Patricia Esparza
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana UniversityIndianapolisIndianaUSA
| | - Abigail Pajulas
- Department of Microbiology and ImmunologyIndiana UniversityIndianapolisIndianaUSA
| | - Jocelyn Wang
- Department of Microbiology and ImmunologyIndiana UniversityIndianapolisIndianaUSA
| | - Rakshin Kharwadkar
- Department of Microbiology and ImmunologyIndiana UniversityIndianapolisIndianaUSA
| | - Hongyu Gao
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Mark H. Kaplan
- Department of Microbiology and ImmunologyIndiana UniversityIndianapolisIndianaUSA
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana UniversityIndianapolisIndianaUSA
| | - Robert S. Tepper
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
15
|
Li J, Ruggiero-Ruff RE, He Y, Qiu X, Lainez NM, Villa PA, Godzik A, Coss D, Nair MG. Sexual dimorphism in obesity is governed by RELMα regulation of adipose macrophages and eosinophils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523880. [PMID: 36711654 PMCID: PMC9882128 DOI: 10.1101/2023.01.13.523880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Obesity incidence is increasing worldwide with the urgent need to identify new therapeutics. Sex differences in immune cell activation drive obesity-mediated pathologies where males are more susceptible to obesity co-morbidities and exacerbated inflammation. Here, we demonstrate that the macrophage-secreted protein RELMα critically protects females against high fat diet-induced obesity. Compared to male mice, RELMα levels were elevated in both control and high fat dietfed females and correlated with adipose macrophages and eosinophils. RELMα-deficient females gained more weight and had pro-inflammatory macrophage accumulation and eosinophil loss, while both RELMα treatment and eosinophil transfer rescued this phenotype. Single cell RNA-sequencing of the adipose stromal vascular fraction was performed and identified sex and RELMα-dependent changes. Genes involved in oxygen sensing and iron homeostasis, including hemoglobin and lncRNA Gm47283, correlated with increased obesity, while eosinophil chemotaxis and response to amyloid-beta were protective. Monocyte-to-macrophage transition was also dysregulated in RELMα-deficient animals. Collectively, these studies implicate a RELMα-macrophage-eosinophil axis in sex-specific protection against obesity and uncover new therapeutic targets for obesity.
Collapse
Affiliation(s)
- Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Rebecca E. Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Yuxin He
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Xinru Qiu
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California Riverside, Riverside, CA, USA
| | - Nancy M. Lainez
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Pedro A. Villa
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
16
|
A homotetrameric hemoglobin expressed in alveolar epithelial cells increases blood oxygenation in high-altitude plateau pika (Ochotona curzoniae). Cell Rep 2022; 41:111446. [DOI: 10.1016/j.celrep.2022.111446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/28/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
|
17
|
Lin Z, Sui X, Jiao W, Chen C, Zhang X, Zhao J. Mechanism investigation and experiment validation of capsaicin on uterine corpus endometrial carcinoma. Front Pharmacol 2022; 13:953874. [PMID: 36210802 PMCID: PMC9532580 DOI: 10.3389/fphar.2022.953874] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Using bioinformatics analysis and experimental operations, we intend to analyze the potential mechanism of action of capsaicin target gene GATA1 in the treatment of uterine corpus endometrial carcinoma (UCEC) and develop a prognostic model for the disease to validate this model. Methods: By obtaining capsaicin and UCEC-related DR-DEGs, the prognosis-related gene GATA1 was screened. The survival analysis was conducted via establishing high and low expression groups of GATA1. Whether the GATA1 could be an independent prognostic factor for UCEC, it was also validated. The therapeutic mechanism of capsaicin-related genes in UCEC was further investigated using enrichment analysis and immune methods as well as in combination with single-cell sequencing data. Finally, it was validated by cell experiments. Results: GATA1, a high-risk gene associated with prognosis, was obtained by screening. Kaplan-Meier analysis showed that the survival of the high expression group was lower than that of low expression group. ROC curves showed that the prediction effect of the model was good and stable (1-year area under curve (AUC): 0.601; 2-years AUC: 0.575; 3-years AUC: 0.610). Independent prognosis analysis showed that the GATA1 can serve as an independent prognostic factor for UCEC. Enrichment analysis showed that “neuroactive Ligand - receptor interaction and TYPE I DIABETES MELLITUS” had a significant enrichment effect. Single-cell sequencing showed that the GATA1 was significantly expressed in mast cells. Cell experiments showed that the capsaicin significantly reduced the UCEC cell activity and migration ability, as well as inhibited the expression of GATA1. Conclusion: This study suggests that the capsaicin has potential value and application prospect in the treatment of UCEC. It provides new genetic markers for the prognosis of UCEC patients.
Collapse
Affiliation(s)
- Zhiheng Lin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjian Jiao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chong Chen
- Obstetrics Department of Affiliated Hospital of Weifang Medical College, Weifang, China
| | - Xiaodan Zhang
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Junde Zhao, ; Xiaodan Zhang,
| | - Junde Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University Cheeloo College of Medicine Laboratory of Basic Medical Sciences, Jinan, China
- *Correspondence: Junde Zhao, ; Xiaodan Zhang,
| |
Collapse
|
18
|
Iarovaia OV, Ulianov SV, Ioudinkova ES, Razin SV. Segregation of α- and β-Globin Gene Cluster in Vertebrate Evolution: Chance or Necessity? BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1035-1049. [PMID: 36180994 DOI: 10.1134/s0006297922090140] [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: 07/22/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
The review is devoted to the patterns of evolution of α- and β-globin gene domains. A hypothesis is presented according to which segregation of the ancestral cluster of α/β-globin genes in Amniota occurred due to the performance by α-globins and β-globins of non-canonical functions not related to oxygen transport.
Collapse
Affiliation(s)
- Olga V Iarovaia
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Sergey V Ulianov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Elena S Ioudinkova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Sergey V Razin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
19
|
Zheng R, Yan Y, Pu J, Zhang B. Physiological and Pathological Functions of Neuronal Hemoglobin: A Key Underappreciated Protein in Parkinson's Disease. Int J Mol Sci 2022; 23:9088. [PMID: 36012351 PMCID: PMC9408843 DOI: 10.3390/ijms23169088] [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: 06/29/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
The expression of Hemoglobin (Hb) is not restricted to erythrocytes but is also present in neurons. Hb is selectively enriched in vulnerable mesencephalic dopaminergic neurons of Parkinson's disease (PD) instead of resistant neurons. Controversial results of neuronal Hb levels have been reported in postmortem brains of PD patients: although neuronal Hb levels may decline in PD patients, elderly men with higher Hb levels have an increased risk of developing PD. α-synuclein, a key protein involved in PD pathology, interacts directly with Hb protein and forms complexes in erythrocytes and brains of monkeys and humans. These complexes increase in erythrocytes and striatal cytoplasm, while they decrease in striatal mitochondria with aging. Besides, the colocalization of serine 129-phosphorylated (Pser129) α-synuclein and Hb β chains have been found in the brains of PD patients. Several underlying molecular mechanisms involving mitochondrial homeostasis, α-synuclein accumulation, iron metabolism, and hormone-regulated signaling pathways have been investigated to assess the relationship between neuronal Hb and PD development. The formation of fibrils with neuronal Hb in various neurodegenerative diseases may indicate a common fibrillization pathway and a widespread target that could be applied in neurodegeneration therapy.
Collapse
Affiliation(s)
| | | | - Jiali Pu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
20
|
Lu Y, Wang J, Tang F, Pratap UP, Sareddy GR, Dhandapani KM, Capuano A, Arvanitakis Z, Vadlamudi RK, Brann DW. Regulation and Role of Neuron-Derived Hemoglobin in the Mouse Hippocampus. Int J Mol Sci 2022; 23:5360. [PMID: 35628182 PMCID: PMC9140924 DOI: 10.3390/ijms23105360] [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: 04/20/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022] Open
Abstract
Hemoglobin (Hb) is the oxygen transport protein in erythrocytes. In blood, Hb is a tetramer consisting of two Hb-alpha (Hb-α) chains and two Hb-beta (Hb-β) chains. A number of studies have also shown that Hb-α is also expressed in neurons in both the rodent and human brain. In the current study, we examined for age-related regulation of neuronal Hb-α and hypoxia in the hippocampus and cerebral cortex of intact male and female mice. In addition, to confirm the role and functions of neuronal Hb-α, we also utilized lentivirus CRISPR interference-based Hb-α knockdown (Hb-α CRISPRi KD) in the non-ischemic and ischemic mouse hippocampus and examined the effect on neuronal oxygenation, as well as induction of hypoxia-inducible factor-1α (HIF-1α) and its downstream pro-apoptotic factors, PUMA and NOXA, and on neuronal survival and neurodegeneration. The results of the study revealed an age-related decrease in neuronal Hb-α levels and correlated increase in hypoxia in the hippocampus and cortex of intact male and female mice. Sex differences were observed with males having higher neuronal Hb-α levels than females in all brain regions at all ages. In vivo Hb-α CRISPRi KD in the mouse hippocampus resulted in increased hypoxia and elevated levels of HIF-1α, PUMA and NOXA in the non-ischemic and ischemic mouse hippocampus, effects that were correlated with a significant decrease in neuronal survival and increased neurodegeneration. As a whole, these findings indicate that neuronal Hb-α decreases with age in mice and has an important role in regulating neuronal oxygenation and neuroprotection.
Collapse
Affiliation(s)
- Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (Y.L.); (K.M.D.)
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| | - Fulei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Krishnan M. Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (Y.L.); (K.M.D.)
| | - Ana Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA; (A.C.); (Z.A.)
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA; (A.C.); (Z.A.)
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Darrell W. Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| |
Collapse
|
21
|
Keller TCS, Lechauve C, Keller AS, Brooks S, Weiss MJ, Columbus L, Ackerman H, Cortese-Krott MM, Isakson BE. The role of globins in cardiovascular physiology. Physiol Rev 2022; 102:859-892. [PMID: 34486392 PMCID: PMC8799389 DOI: 10.1152/physrev.00037.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Globin proteins exist in every cell type of the vasculature, from erythrocytes to endothelial cells, vascular smooth muscle cells, and peripheral nerve cells. Many globin subtypes are also expressed in muscle tissues (including cardiac and skeletal muscle), in other organ-specific cell types, and in cells of the central nervous system (CNS). The ability of each of these globins to interact with molecular oxygen (O2) and nitric oxide (NO) is preserved across these contexts. Endothelial α-globin is an example of extraerythrocytic globin expression. Other globins, including myoglobin, cytoglobin, and neuroglobin, are observed in other vascular tissues. Myoglobin is observed primarily in skeletal muscle and smooth muscle cells surrounding the aorta or other large arteries. Cytoglobin is found in vascular smooth muscle but can also be expressed in nonvascular cell types, especially in oxidative stress conditions after ischemic insult. Neuroglobin was first observed in neuronal cells, and its expression appears to be restricted mainly to the CNS and the peripheral nervous system. Brain and CNS neurons expressing neuroglobin are positioned close to many arteries within the brain parenchyma and can control smooth muscle contraction and thus tissue perfusion and vascular reactivity. Overall, reactions between NO and globin heme iron contribute to vascular homeostasis by regulating vasodilatory NO signals and scavenging reactive species in cells of the mammalian vascular system. Here, we discuss how globin proteins affect vascular physiology, with a focus on NO biology, and offer perspectives for future study of these functions.
Collapse
Affiliation(s)
- T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Steven Brooks
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, Virginia
| | - Hans Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
22
|
Li J, An Z, Wei L, Xu B, Wang Z, Gao C, Wei L, Qi D, Shi P, Zhang T, Wei D. A New Homotetramer Hemoglobin in the Pulmonary Surfactant of Plateau Zokors (Myospalax Baileyi). Front Genet 2022; 13:824049. [PMID: 35368669 PMCID: PMC8967358 DOI: 10.3389/fgene.2022.824049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
The plateau zokor (Myospalax baileyi) is a native species to the Qinghai-Tibetan Plateau, inhabiting hypoxia and hypercapnia sealed subterranean burrows that pose several unique physiological challenges. In this study, we observed a novel heme-containing protein in the pulmonary surfactant (PS) of plateau zokor, identified the encoding gene of the protein, predicted its origination and structure, verified its expression in alveolar epithelial cells, and determined the protein’s affinity to oxygen and its effect on the oxygen-dissolving capability in the PS of plateau zokors. The protein is an unusual homotetramer hemoglobin consisting of four γ-like subunits, and the subunit is encoded by a paralog gene of γ, that is γ-like. The divergence time of γ-like from γ is estimated by the molecular clock to be about 2.45 Mya. The generation of γ-like in plateau zokors might well relate to long-time stress of the high land hypoxia. Unlike γ, the γ-like has a hypoxia response element (HRE) and a lung tissue-specific enhancer in its upstream region, and it is expressed specifically in lung tissues and up-regulated by hypoxia. The protein is named as γ4-like which is expressed specifically in Alveolar epithelial type II (ATII) cells and secreted into the alveolar cavities through the osmiophilic multilamellar body (LBs). The γ4-like has a higher affinity to oxygen, and that increases significantly oxygen-dissolving capability in the PS of plateau zokors by its oxygenation function, which might be beneficial for the plateau zokors to obtain oxygen from the severe hypoxia environments by facilitating oxygen diffusion from alveoli to blood.
Collapse
Affiliation(s)
- Jimei Li
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
| | - Zhifang An
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Linna Wei
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Bo Xu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Zhijie Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Conghui Gao
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Lian Wei
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Delin Qi
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Dengbang Wei, ; Tongzuo Zhang, ; Peng Shi,
| | - Tongzuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- *Correspondence: Dengbang Wei, ; Tongzuo Zhang, ; Peng Shi,
| | - Dengbang Wei
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- *Correspondence: Dengbang Wei, ; Tongzuo Zhang, ; Peng Shi,
| |
Collapse
|
23
|
Gardner PR. Ordered Motions in the Nitric-Oxide Dioxygenase Mechanism of Flavohemoglobin and Assorted Globins with Tightly Coupled Reductases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1414:45-96. [PMID: 36520413 DOI: 10.1007/5584_2022_751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nitric-oxide dioxygenases (NODs) activate and combine O2 with NO to form nitrate. A variety of oxygen-binding hemoglobins with associated partner reductases or electron donors function as enzymatic NODs. Kinetic and structural investigations of the archetypal two-domain microbial flavohemoglobin-NOD have illuminated an allosteric mechanism that employs selective tunnels for O2 and NO, gates for NO and nitrate, transient O2 association with ferric heme, and an O2 and NO-triggered, ferric heme spin crossover-driven, motion-controlled, and dipole-regulated electron-transfer switch. The proposed mechanism facilitates radical-radical coupling of ferric-superoxide with NO to form nitrate while preventing suicidal ferrous-NO formation. Diverse globins display the structural and functional motifs necessary for a similar allosteric NOD mechanism. In silico docking simulations reveal monomeric erythrocyte hemoglobin alpha-chain and beta-chain intrinsically matched and tightly coupled with NADH-cytochrome b5 oxidoreductase and NADPH-cytochrome P450 oxidoreductase, respectively, forming membrane-bound flavohemoglobin-like mammalian NODs. The neuroprotective neuroglobin manifests a potential NOD role in a close-fitting ternary complex with membrane-bound NADH-cytochrome b5 oxidoreductase and cytochrome b5. Cytoglobin interfaces weakly with cytochrome b5 for O2 and NO-regulated electron-transfer and coupled NOD activity. The mechanistic model also provides insight into the evolution of O2 binding cooperativity in hemoglobin and a basis for the discovery of allosteric NOD inhibitors.
Collapse
|
24
|
Erlandsson L, Masoumi Z, Hansson LR, Hansson SR. The roles of free iron, heme, haemoglobin, and the scavenger proteins haemopexin and alpha-1-microglobulin in preeclampsia and fetal growth restriction. J Intern Med 2021; 290:952-968. [PMID: 34146434 DOI: 10.1111/joim.13349] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a complex pregnancy syndrome characterised by maternal hypertension and organ damage after 20 weeks of gestation and is associated with an increased risk of cardiovascular disease later in life. Extracellular haemoglobin (Hb) and its metabolites heme and iron are highly toxic molecules and several defence mechanisms have evolved to protect the tissue. OBJECTIVES We will discuss the roles of free iron, heme, Hb, and the scavenger proteins haemopexin and alpha-1-microglobulin in pregnancies complicated by PE and fetal growth restriction (FGR). CONCLUSION In PE, oxidative stress causes syncytiotrophoblast (STB) stress and increased shedding of placental STB-derived extracellular vesicles (STBEV). The level in maternal circulation correlates with the severity of hypertension and supports the involvement of STBEVs in causing maternal symptoms in PE. In PE and FGR, iron homeostasis is changed, and iron levels significantly correlate with the severity of the disease. The normal increase in plasma volume taking place during pregnancy is less for PE and FGR and therefore have a different impact on, for example, iron concentration, compared to normal pregnancy. Excess iron promotes ferroptosis is suggested to play a role in trophoblast stress and lipotoxicity. Non-erythroid α-globin regulates vasodilation through the endothelial nitric oxide synthase pathway, and hypoxia-induced α-globin expression in STBs in PE placentas is suggested to contribute to hypertension in PE. Underlying placental pathology in PE with and without FGR might be amplified by iron and heme overload causing oxidative stress and ferroptosis. As the placenta becomes stressed, the release of STBEVs increases and affects the maternal vasculature.
Collapse
Affiliation(s)
- Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zahra Masoumi
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lucas R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Obstetrics and Gynecology, Skåne University Hospital, Lund/Malmö, Sweden
| |
Collapse
|
25
|
Marozkina N, Smith L, Zhao Y, Zein J, Chmiel JF, Kim J, Kiselar J, Davis MD, Cunningham RS, Randell SH, Gaston B. Somatic cell hemoglobin modulates nitrogen oxide metabolism in the human airway epithelium. Sci Rep 2021; 11:15498. [PMID: 34326365 PMCID: PMC8322277 DOI: 10.1038/s41598-021-94782-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/09/2021] [Indexed: 12/02/2022] Open
Abstract
Endothelial hemoglobin (Hb)α regulates endothelial nitric oxide synthase (eNOS) biochemistry. We hypothesized that Hb could also be expressed and biochemically active in the ciliated human airway epithelium. Primary human airway epithelial cells, cultured at air–liquid interface (ALI), were obtained by clinical airway brushings or from explanted lungs. Human airway Hb mRNA data were from publically available databases; or from RT-PCR. Hb proteins were identified by immunoprecipitation, immunoblot, immunohistochemistry, immunofluorescence and liquid chromatography- mass spectrometry. Viral vectors were used to alter Hbβ expression. Heme and nitrogen oxides were measured colorimetrically. Hb mRNA was expressed in human ciliated epithelial cells. Heme proteins (Hbα, β, and δ) were detected in ALI cultures by several methods. Higher levels of airway epithelial Hbβ gene expression were associated with lower FEV1 in asthma. Both Hbβ knockdown and overexpression affected cell morphology. Hbβ and eNOS were apically colocalized. Binding heme with CO decreased extracellular accumulation of nitrogen oxides. Human airway epithelial cells express Hb. Higher levels of Hbβ gene expression were associated with airflow obstruction. Hbβ and eNOS were colocalized in ciliated cells, and heme affected oxidation of the NOS product. Epithelial Hb expression may be relevant to human airways diseases.
Collapse
Affiliation(s)
- Nadzeya Marozkina
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA.
| | - Laura Smith
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| | - Yi Zhao
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| | - Joe Zein
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James F Chmiel
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| | - Jeeho Kim
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - Michael D Davis
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| | - Rebekah S Cunningham
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| | - Scott H Randell
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Benjamin Gaston
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, Indiana University School of Medicine, 1044 W. Walnut Street, R4-474, Indianapolis, IN, 46202, USA
| |
Collapse
|
26
|
Ma H, Liu J, Li Z, Xiong H, Zhang Y, Song Y, Lai J. Expression profile analysis reveals hub genes that are associated with immune system dysregulation in primary myelofibrosis. ACTA ACUST UNITED AC 2021; 26:478-490. [PMID: 34238135 DOI: 10.1080/16078454.2021.1945237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTION Primary myelofibrosis (PMF) is a familiar chronic myeloproliferative disease with an unfavorable prognosis. The effect of infection on the prognosis of patients with PMF is crucial. Immune system dysregulation plays a central role in the pathophysiology of PMF. To date, very little research has been conducted on the molecular mechanism of immune compromise in patients with PMF. METHODS To explore potential candidate genes, microarray datasets GSE61629 and 26049 were obtained from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) between PMF patients and normal individuals were evaluated, gene function was measured and a series of hub genes were identified. Several significant immune cells were selected via cell type enrichment analysis. The correlation between hub genes and significant immune cells was determined. RESULTS A total of 282 DEGs were found, involving 217 upregulated genes and 65 downregulated genes. Several immune cells were found to be reduced in PMF, such as CD4+ T cells, CD4+ Tems, CD4+ memory T cells. Gene Ontology (GO) enrichment analysis of DEGs reflected that most biological processes were associated with immune processes. Six hub genes, namely, HP, MPO, MMP9, EPB42, SLC4A1, and ALAS2, were identified, and correlation analysis revealed that these hub genes have a negative correlation with immune cell abundance. CONCLUSIONS Taken together, the gene expression profile of whole blood cells in PMF patients indicated a battery of immune events, and the DEGs and hub genes might contribute to immune system dysregulation.
Collapse
Affiliation(s)
- Haotian Ma
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jincen Liu
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zilong Li
- College of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Huaye Xiong
- College of Resources and Environment, Southwest University, Chongqing, People's Republic of China
| | - Yulei Zhang
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanping Song
- Institute of Hematology, Central Hospital of Xi'an, Xi'an, People's Republic of China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
27
|
Increased susceptibility to dextran sulfate-induced mucositis of iron-overload β-thalassemia mice, another endogenous cause of septicemia in thalassemia. Clin Sci (Lond) 2021; 135:1467-1486. [PMID: 34131711 PMCID: PMC8209035 DOI: 10.1042/cs20210328] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 12/28/2022]
Abstract
Enterocyte damage and gut dysbiosis are caused by iron-overload in thalassemia (Thl), possibly making the gut vulnerable to additional injury. Hence, iron-overload in the heterozygous β-globin deficient (Hbbth3/+) mice were tested with 3% dextran sulfate solution (DSS). With 4 months of iron-gavage, iron accumulation, gut-leakage (fluorescein isothiocyanate dextran (FITC-dextran), endotoxemia, and tight junction injury) in Thl mice were more prominent than WT mice. Additionally, DSS-induced mucositis in iron-overloaded mice from Thl group was also more severe than the WT group as indicated by mortality, liver enzyme, colon injury (histology and tissue cytokines), serum cytokines, and gut-leakage (FITC-dextran, endotoxemia, bacteremia, and the detection of Green-Fluorescent Producing Escherichia coli in the internal organs after an oral administration). However, Lactobacillus rhamnosus GG attenuated the disease severity of DSS in iron-overloaded Thl mice as indicated by mortality, cytokines (colon tissue and serum), gut-leakage (FITC-dextran, endotoxemia, and bacteremia) and fecal dysbiosis (microbiome analysis). Likewise, Lactobacillus conditioned media (LCM) decreased inflammation (supernatant IL-8 and cell expression of TLR-4, nuclear factor κB (NFκB), and cyclooxygenase-2 (COX-2)) and increased transepithelial electrical resistance (TEER) in enterocytes (Caco-2 cells) stimulated by lipopolysaccharide (LPS) and LPS plus ferric ion. In conclusion, in the case of iron-overloaded Thl, there was a pre-existing intestinal injury that wask more vulnerable to DSS-induced bacteremia (gut translocation). Hence, the prevention of gut-derived bacteremia and the monitoring on gut-leakage might be beneficial in patients with thalassemia.
Collapse
|
28
|
Hypoxia-Induced Alpha-Globin Expression in Syncytiotrophoblasts Mimics the Pattern Observed in Preeclamptic Placentas. Int J Mol Sci 2021; 22:ijms22073357. [PMID: 33806017 PMCID: PMC8036899 DOI: 10.3390/ijms22073357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy disorder associated with placental dysfunction and elevated fetal hemoglobin (HbF). Early in pregnancy the placenta harbors hematopoietic stem and progenitor cells (HSPCs) and is an extramedullary source of erythropoiesis. However, globin expression is not unique to erythroid cells and can be triggered by hypoxia. To investigate the role of the placenta in increasing globin levels previously reported in PE, flow cytometry, histological and immunostaining and in situ analyses were used on placenta samples and ex vivo explant cultures. Our results indicated that in PE pregnancies, placental HSPC homing and erythropoiesis were not affected. Non-erythroid alpha-globin mRNA and protein, but not gamma-globin, were detected in syncytiotrophoblasts and stroma of PE placenta samples. Similarly, alpha-globin protein and mRNA were upregulated in normal placenta explants cultured in hypoxia. The upregulation was independent of HIF1 and NRF2, the two main candidates of globin transcription in non-erythroid cells. Our study is the first to demonstrate alpha-globin mRNA expression in syncytiotrophoblasts in PE, induced by hypoxia. However, gamma-globin was only expressed in erythrocytes. We conclude that alpha-globin, but not HbF, is expressed in placental syncytiotrophoblasts in PE and may contribute to the pathology of the disease.
Collapse
|
29
|
Sae-Khow K, Charoensappakit A, Visitchanakun P, Saisorn W, Svasti S, Fucharoen S, Leelahavanichkul A. Pathogen-Associated Molecules from Gut Translocation Enhance Severity of Cecal Ligation and Puncture Sepsis in Iron-Overload β-Thalassemia Mice. J Inflamm Res 2020; 13:719-735. [PMID: 33116751 PMCID: PMC7569041 DOI: 10.2147/jir.s273329] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/12/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Systemic inflammation induced by gut translocation of lipopolysaccharide (LPS), a major component of Gram-negative bacteria, in thalassemia with iron-overload worsens sepsis. However, the impact of (1→3)-β-D-glucan (BG), a major fungal molecule, in iron-overload thalassemia is still unclear. Hence, the influence of BG was explored in 1) iron-overload mice with sepsis induced by cecal ligation and puncture (CLP) surgery; and 2) in bone marrow-derived macrophages (BMMs). Methods The heterozygous β-globin-deficient mice, Hbbth3/+ mice, were used as representative thalassemia (TH) mice. Iron overload was generated by 6 months of oral iron administration before CLP surgery- induced sepsis in TH mice and wild-type (WT) mice. Additionally, BMMs from both mouse strains were used to explore the impact of BG. Results Without sepsis, iron-overload TH mice demonstrated more severe intestinal mucosal injury (gut leakage) with higher LPS and BG in serum, from gut translocation, when compared with WT mice. With CLP in iron-overload mice, sepsis severity in TH mice was more severe than WT as determined by survival analysis, organ injury (kidney and liver), bacteremia, endotoxemia, gut leakage (FITC-dextran) and serum BG. Activation by LPS plus BG (LPS+BG) in BMMs and in peripheral blood-derived neutrophils (both WT and TH cells) demonstrated more prominent cytokine production when compared with LPS activation alone. In parallel, LPS+BG also prominently induced genes expression of M1 macrophage polarization (iNOS, TNF-α and IL-1β) in both WT and TH cells in comparison with LPS activation alone. In addition, LPS+BG activated macrophage cytokine production was enhanced by a high dose of ferric ion (800 mM), more predominantly in TH macrophages compared with WT cells. Moreover, LPS+BG induced higher glycolysis activity with similar respiratory capacity in RAW264.7 (a macrophage cell line) compared with LPS activation alone. These data support an additive pro-inflammatory effect of BG upon LPS. Conclusion The enhanced-severity of sepsis in iron-overload TH mice was due to 1) increased LPS and BG in serum from iron-induced gut-mucosal injury; and 2) the pro-inflammatory amplification by ferric ion on LPS+BG activation.
Collapse
Affiliation(s)
- Kritsanawan Sae-Khow
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Awirut Charoensappakit
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Peerapat Visitchanakun
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Wilasinee Saisorn
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | - Asada Leelahavanichkul
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand.,Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
30
|
Lim M, Brown HM, Kind KL, Thompson JG, Dunning KR. Hemoglobin: potential roles in the oocyte and early embryo†. Biol Reprod 2020; 101:262-270. [PMID: 31058953 DOI: 10.1093/biolre/ioz078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/02/2019] [Accepted: 05/04/2019] [Indexed: 12/13/2022] Open
Abstract
Hemoglobin (Hb) is commonly known for its capacity to bind and transport oxygen and carbon dioxide in erythroid cells. However, it plays additional roles in cellular function and health due to its capacity to bind other gases including nitric oxide. Further, Hb acts as a potent antioxidant, quenching reactive oxygen species. Despite its potential roles in cellular function, the preponderance of Hb research remains focused on its role in oxygen regulation. There is increasing evidence that Hb expression is more ubiquitous than previously thought, with Hb and its variants found in a myriad of cell types ranging from macrophages to spermatozoa. The majority of nonerythroid cell types that express Hb are situated within hypoxic environments, suggesting Hb may play a role in hypoxia-inducible factor-regulated gene expression by controlling the level of oxygen available or as an adaptation to low oxygen providing a mechanism to store oxygen. Oocyte maturation and preimplantation embryo development occur within the low oxygen environments of the antral follicle and oviduct/uterus, respectively. Interestingly, Hb was recently found in human cumulus and granulosa cells and murine cumulus-oocyte complexes and preimplantation embryos. Here, we consolidate and analyze the research generated todate on Hb expression in nonerythroid cells with a particular focus on reproductive cell types. We outline future directions of this research to elucidate the role of Hb during oocyte maturation and preimplantation embryo development and finally, we explore the potential clinical applications and benefits of Hb supplementation during the in vitro culture of gametes and embryos.
Collapse
Affiliation(s)
- Megan Lim
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| | - Hannah M Brown
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Karen L Kind
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jeremy G Thompson
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kylie R Dunning
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Australian Research Council Centre of Excellence for Nanoscale BioPhotonics.,Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
31
|
Lessons from the post-genomic era: Globin diversity beyond oxygen binding and transport. Redox Biol 2020; 37:101687. [PMID: 32863222 PMCID: PMC7475203 DOI: 10.1016/j.redox.2020.101687] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Vertebrate hemoglobin (Hb) and myoglobin (Mb) were among the first proteins whose structures and sequences were determined over 50 years ago. In the subsequent pregenomic period, numerous related proteins came to light in plants, invertebrates and bacteria, that shared the myoglobin fold, a signature sequence motif characteristic of a 3-on-3 α-helical sandwich. Concomitantly, eukaryote and bacterial globins with a truncated 2-on-2 α-helical fold were discovered. Genomic information over the last 20 years has dramatically expanded the list of known globins, demonstrating their existence in a limited number of archaeal genomes, a majority of bacterial genomes and an overwhelming majority of eukaryote genomes. In vertebrates, 6 additional globin types were identified, namely neuroglobin (Ngb), cytoglobin (Cygb), globin E (GbE), globin X (GbX), globin Y (GbY) and androglobin (Adgb). Furthermore, functions beyond the familiar oxygen transport and storage have been discovered within the vertebrate globin family, including NO metabolism, peroxidase activity, scavenging of free radicals, and signaling functions. The extension of the knowledge on globin functions suggests that the original roles of bacterial globins must have been enzymatic, involved in defense against NO toxicity, and perhaps also as sensors of O2, regulating taxis away or towards high O2 concentrations. In this review, we aimed to discuss the evolution and remarkable functional diversity of vertebrate globins with particular focus on the variety of non-canonical expression sites of mammalian globins and their according impressive variability of atypical functions.
Collapse
|
32
|
Lipid-Protein and Protein-Protein Interactions in the Pulmonary Surfactant System and Their Role in Lung Homeostasis. Int J Mol Sci 2020; 21:ijms21103708. [PMID: 32466119 PMCID: PMC7279303 DOI: 10.3390/ijms21103708] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary surfactant is a lipid/protein complex synthesized by the alveolar epithelium and secreted into the airspaces, where it coats and protects the large respiratory air–liquid interface. Surfactant, assembled as a complex network of membranous structures, integrates elements in charge of reducing surface tension to a minimum along the breathing cycle, thus maintaining a large surface open to gas exchange and also protecting the lung and the body from the entrance of a myriad of potentially pathogenic entities. Different molecules in the surfactant establish a multivalent crosstalk with the epithelium, the immune system and the lung microbiota, constituting a crucial platform to sustain homeostasis, under health and disease. This review summarizes some of the most important molecules and interactions within lung surfactant and how multiple lipid–protein and protein–protein interactions contribute to the proper maintenance of an operative respiratory surface.
Collapse
|
33
|
Visitchanakun P, Saisorn W, Wongphoom J, Chatthanathon P, Somboonna N, Svasti S, Fucharoen S, Leelahavanichkul A. Gut leakage enhances sepsis susceptibility in iron-overloaded β-thalassemia mice through macrophage hyperinflammatory responses. Am J Physiol Gastrointest Liver Physiol 2020; 318:G966-G979. [PMID: 32308038 DOI: 10.1152/ajpgi.00337.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Iron overload induces intestinal-permeability defect (gut leakage), and gut translocation of organismal molecules might enhance systemic inflammation and sepsis severity in patients with thalassemia (Thal). Hence, iron administration in Hbbth3/+ mice, heterozygous β-globin-deficient Thal mice, was explored. Oral iron administration induced more severe secondary hemochromatosis and gut leakage in Thal mice compared with wild-type (WT) mice. Gut leakage was determined by 1) FITC-dextran assay, 2) spontaneous serum elevation of endotoxin (LPS) and (1→3)-β-d-glucan (BG), molecular structures of gut-organisms, and 3) reduction of tight-junction molecules with increased enterocyte apoptosis (activated caspase-3) by immunofluorescent staining. Iron overload also enhanced serum cytokines and increased Bacteroides spp. (gram-negative bacteria) in feces as analyzed by microbiome analysis. LPS injection in iron-overloaded Thal mice produced higher mortality and prominent cytokine responses. Additionally, stimulation with LPS plus iron in macrophage from Thal mice induced higher cytokines production with lower β-globin gene expression compared with WT. Furthermore, possible gut leakage as determined by elevated LPS or BG (>60 pg/mL) in serum without systemic infection was demonstrated in 18 out of 41 patients with β-thalassemia major. Finally, enhanced LPS-induced cytokine responses of mononuclear cells from these patients compared with cells from healthy volunteers were demonstrated. In conclusion, oral iron administration in Thal mice induced more severe gut leakage and increased fecal gram-negative bacteria, resulting in higher levels of endotoxemia and serum inflammatory cytokines compared with WT. Preexisting hyperinflammatory cytokines in iron-overloaded Thal enhanced susceptibility toward infection.NEW & NOTEWORTHY Although the impact of iron accumulation in several organs of patients with thalassemia is well known, the adverse effect of iron accumulation in gut is not frequently mentioned. Here, we demonstrated iron-induced gut-permeability defect, impact of organismal molecules from gut translocation of, and macrophage functional defect upon the increased sepsis susceptibility in thalassemia mice.
Collapse
Affiliation(s)
- Peerapat Visitchanakun
- Faculty of Medicine, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand.,Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Wilasinee Saisorn
- Faculty of Medicine, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Jutamas Wongphoom
- Department of Pathology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Piraya Chatthanathon
- Faculty of Science, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Naraporn Somboonna
- Faculty of Science, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand.,Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | - Asada Leelahavanichkul
- Faculty of Medicine, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand.,Translational Research in Inflammation and Immunology Research Unit, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
34
|
Wang X, Tang S, Qin F, Liu Y, Liang Z, Cai H, Mo L, Xiao D, Guo S, Ouyang Y, Sun B, Lu C, Li X. Proteomics and phosphoproteomics study of LCMT1 overexpression and oxidative stress: overexpression of LCMT1 arrests H 2O 2-induced lose of cells viability. Redox Rep 2020; 24:1-9. [PMID: 30898057 PMCID: PMC6748586 DOI: 10.1080/13510002.2019.1595332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objectives: Protein phosphatase 2A (PP2A), a major serine/threonine
phosphatase, is also known to be a target of ROS. The methylation of PP2A can be
catalyzed by leucine carboxyl methyltransferase-1 (LCMT1), which regulates PP2A
activity and substrate specificity. Methods: In the previous study, we have showed that LCMT1-dependent
PP2Ac methylation arrests H2O2-induced cell oxidative
stress damage. To explore the possible protective mechanism, we performed
iTRAQ-based comparative quantitative proteomics and phosphoproteomics studies of
H2O2-treated vector control and LCMT1-overexpressing
cells. Results: A total of 4480 non-redundant proteins and 3801 unique
phosphopeptides were identified by this means. By comparing the
H2O2-regulated proteins in LCMT1-overexpressing and
vector control cells, we found that these differences were mainly related to
protein phosphorylation, gene expression, protein maturation, the cytoskeleton
and cell division. Further investigation of LCMT1 overexpression-specific
regulated proteins under H2O2 treatment supported the idea
that LCMT1 overexpression induced ageneral dephosphorylation of proteins and
indicated increased expression of non-erythrocytic hemoglobin, inactivation of
MAPK3 and regulation of proteins related to Rho signal transduction, which were
known to be linked to the regulation of the cytoskeleton. Discussion: These data provide proteomics and phosphoproteomics
insights into the association of LCMT1-dependent PP2Ac methylation and oxidative
stress and indirectly indicate that the methylation of PP2A plays an important
role against oxidative stress.
Collapse
Affiliation(s)
- Xinhang Wang
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Shen Tang
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Fu Qin
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Yuyang Liu
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Ziwei Liang
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Haiqing Cai
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Laiming Mo
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Deqiang Xiao
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China
| | - Songcao Guo
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China
| | - Yiqiang Ouyang
- d Laboratory Animal Centre , Guangxi Medical University , Nanning , People's Republic of China
| | - Bin Sun
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Cailing Lu
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Xiyi Li
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| |
Collapse
|
35
|
Short-Term Caloric Restriction Attenuates Obesity-Induced Pro-Inflammatory Response in Male Rhesus Macaques. Nutrients 2020; 12:nu12020511. [PMID: 32085416 PMCID: PMC7071433 DOI: 10.3390/nu12020511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (WAT) hypertrophy is an essential hallmark of obesity and is associated with the activation of resident immune cells. While the benefits of caloric restriction (CR) on health span are generally accepted, its effects on WAT physiology are not well understood. We previously demonstrated that short-term CR reverses obesity in male rhesus macaques exposed to a high-fat Western-style diet (WSD). Here, we analyzed subcutaneous WAT biopsies collected from this cohort of animals before and after WSD and following CR. This analysis showed that WSD induced adipocyte hypertrophy and inhibited β-adrenergic-simulated lipolysis. CR reversed adipocyte hypertrophy, but WAT remained insensitive to β-adrenergic agonist stimulation. Whole-genome transcriptional analysis revealed that β3-adrenergic receptor and de novo lipogenesis genes were downregulated by WSD and remained downregulated after CR. In contrast, WSD-induced pro-inflammatory gene expression was effectively reversed by CR. Furthermore, peripheral blood monocytes isolated during the CR period exhibited a significant reduction in the production of pro-inflammatory cytokines compared to those obtained after WSD. Collectively, this study demonstrates that short-term CR eliminates an obesity-induced pro-inflammatory response in WAT and peripheral monocytes.
Collapse
|
36
|
Lim M, Brown HM, Kind KL, Breen J, Anastasi MR, Ritter LJ, Tregoweth EK, Dinh DT, Thompson JG, Dunning KR. Haemoglobin expression in in vivo murine preimplantation embryos suggests a role in oxygen-regulated gene expression. Reprod Fertil Dev 2019; 31:724-734. [PMID: 30482269 DOI: 10.1071/rd17321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/24/2018] [Indexed: 01/17/2023] Open
Abstract
Haemoglobin expression is not restricted to erythroid cells. We investigated the gene expression of the haemoglobin subunits haemoglobin, alpha adult chain 1 (Hba-a1) and haemoglobin, beta (Hbb), 2,3-bisphosphoglycerate mutase (Bpgm) and the oxygen-regulated genes BCL2/adenovirus E1B interacting protein 3 (Bnip3), solute carrier family 2 (facilitated glucose transporter), member 1 (Slc2a1) and N-myc downstream regulated gene 1 (Ndrg1) in the murine preimplantation embryo, comparing invivo to invitro gene expression. Relatively high levels of Hba-a1 and Hbb were expressed invivo from the 2-cell to blastocyst stage; in contrast, little or no expression occurred invitro. We hypothesised that the presence of haemoglobin invivo creates a low oxygen environment to induce oxygen-regulated gene expression, supported by high expression of Slc2a1 and Ndrg1 in invivo relative to invitro embryos. In addition, analysis of an invitro-derived human embryo gene expression public dataset revealed low expression of haemoglobin subunit alpha (HBA) and HBB, and high expression of BPGM. To explore whether there was a developmental stage-specific effect of haemoglobin, we added exogenous haemoglobin either up to the 4-cell stage or throughout development to the blastocyst stage, but observed no difference in blastocyst rate or the inner cell mass to trophectoderm cell ratio. We conclude that haemoglobin in the invivo preimplantation embryo raises an interesting premise of potential mechanisms for oxygen regulation, which may influence oxygen-regulated gene expression.
Collapse
Affiliation(s)
- M Lim
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - H M Brown
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - K L Kind
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - J Breen
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - M R Anastasi
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - L J Ritter
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - E K Tregoweth
- Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5005, Australia
| | - D T Dinh
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - J G Thompson
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - K R Dunning
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| |
Collapse
|
37
|
Greene E, Flees J, Dadgar S, Mallmann B, Orlowski S, Dhamad A, Rochell S, Kidd M, Laurendon C, Whitfield H, Brearley C, Rajaram N, Walk C, Dridi S. Quantum Blue Reduces the Severity of Woody Breast Myopathy via Modulation of Oxygen Homeostasis-Related Genes in Broiler Chickens. Front Physiol 2019; 10:1251. [PMID: 31632293 PMCID: PMC6781743 DOI: 10.3389/fphys.2019.01251] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
The incidence of woody breast (WB) is increasing on a global scale representing a significant welfare problem and economic burden to the poultry industry and for which there is no effective treatment due to its unknown etiology. In this study, using diffuse reflectance spectroscopy (DRS) coupled with iSTAT portable clinical analyzer, we provide evidence that the circulatory- and breast muscle-oxygen homeostasis is dysregulated [low oxygen and hemoglobin (HB) levels] in chickens with WB myopathy compared to healthy counterparts. Molecular analysis showed that blood HB subunit Mu (HBM), Zeta (HBZ), and hephaestin (HEPH) expression were significantly down regulated; however, the expression of the subunit rho of HB beta (HBBR) was upregulated in chicken with WB compared to healthy counterparts. The breast muscle HBBR, HBE, HBZ, and hypoxia-inducible factor prolyl hydroxylase 2 (PHD2) mRNA abundances were significantly down regulated in WB-affected compared to normal birds. The expression of HIF-1α at mRNA and protein levels was significantly induced in breasts of WB-affected compared to unaffected birds confirming a local hypoxic status. The phosphorylated levels of the upstream mediators AKT at Ser473 site, mTOR at Ser2481 site, and PI3K P85 at Tyr458 site, as well as their mRNA levels were significantly increased in breasts of WB-affected birds. In attempt to identify a nutritional strategy to reduce WB incidence, male broiler chicks (Cobb 500, n = 576) were randomly distributed into 48 floor pens and subjected to six treatments (12 birds/pen; 8 pens/treatment): a nutrient adequate control group (PC), the PC supplemented with 0.3% myo-inositol (PC + MI), a negative control (NC) deficient in available P and Ca by 0.15 and 0.16%, respectively, the NC fed with quantum blue (QB) at 500 (NC + 500 FTU), 1,000 (NC + 1,000 FTU), or 2,000 FTU/kg of feed (NC + 2,000 FTU). Although QB-enriched diets did not affect growth performances (FCR and FE), it did reduce the severity of WB by 5% compared to the PC diet. This effect is mediated by reversing the expression profile of oxygen homeostasis-related genes; i.e., significant down regulation of HBBR and upregulation of HBM, HBZ, and HEPH in blood, as well as a significant upregulation of HBA1, HBBR, HBE, HBZ, and PHD2 in breast muscle compared to the positive control.
Collapse
Affiliation(s)
- Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Joshua Flees
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sina Dadgar
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Barbara Mallmann
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sara Orlowski
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Ahmed Dhamad
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Samuel Rochell
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Michael Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Caroline Laurendon
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Hayley Whitfield
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Charles Brearley
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Narasimhan Rajaram
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | | | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
38
|
Leite GGF, Scicluna BP, van der Poll T, Salomão R. Genetic signature related to heme-hemoglobin metabolism pathway in sepsis secondary to pneumonia. NPJ Syst Biol Appl 2019; 5:26. [PMID: 31396396 PMCID: PMC6672010 DOI: 10.1038/s41540-019-0105-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated inflammatory response to pathogens. Bioinformatics and transcriptomics studies contribute to get a better understanding of the pathogenesis of sepsis. These studies revealed differentially expressed genes (DEGs) in sepsis involved in several pathways. Here we investigated the gene expression profiles of blood leukocytes using three microarray datasets of sepsis secondary to pneumonia, focusing on the heme/hemoglobin metabolism pathway. We demonstrate that the heme/hemoglobin metabolism pathway was found to be enriched in these three cohorts with four common genes (ALAS2, AHSP, HBD, and CA1). Several studies show that these four genes are involved in the cytoprotection of non-erythrocyte cells in response to different stress conditions. The upregulation of heme/hemoglobin metabolism in sepsis might be a protective response of white cells to the hostile environment present in septic patients (follow-up samples).
Collapse
Affiliation(s)
- Giuseppe Gianini Figuerêido Leite
- Division of Infectious Diseases, Department of Medicine, Hospital São Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Brendon P. Scicluna
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Center of Infection and Immunity Amsterdam, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Center of Infection and Immunity Amsterdam, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Reinaldo Salomão
- Division of Infectious Diseases, Department of Medicine, Hospital São Paulo, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Zhao FL, Ahn JJ, Chen ELY, Yi TJ, Stickle NH, Spaner D, Zúñiga-Pflücker JC, Dunn SE. Peroxisome Proliferator-Activated Receptor-δ Supports the Metabolic Requirements of Cell Growth in TCRβ-Selected Thymocytes and Peripheral CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:2664-2682. [PMID: 30257885 DOI: 10.4049/jimmunol.1800374] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022]
Abstract
During T cell development, progenitor thymocytes undergo a large proliferative burst immediately following successful TCRβ rearrangement, and defects in genes that regulate this proliferation have a profound effect on thymus cellularity and output. Although the signaling pathways that initiate cell cycling and nutrient uptake after TCRβ selection are understood, less is known about the transcriptional programs that regulate the metabolic machinery to promote biomass accumulation during this process. In this article, we report that mice with whole body deficiency in the nuclear receptor peroxisome proliferator-activated receptor-δ (PPARδmut) exhibit a reduction in spleen and thymus cellularity, with a decrease in thymocyte cell number starting at the double-negative 4 stage of thymocyte development. Although in vivo DNA synthesis was normal in PPARδmut thymocytes, studies in the OP9-delta-like 4 in vitro system of differentiation revealed that PPARδmut double-negative 3 cells underwent fewer cell divisions. Naive CD4+ T cells from PPARδmut mice also exhibited reduced proliferation upon TCR and CD28 stimulation in vitro. Growth defects in PPAR-δ-deficient thymocytes and peripheral CD4+ T cells correlated with decreases in extracellular acidification rate, mitochondrial reserve, and expression of a host of genes involved in glycolysis, oxidative phosphorylation, and lipogenesis. By contrast, mice with T cell-restricted deficiency of Ppard starting at the double-positive stage of thymocyte development, although exhibiting defective CD4+ T cell growth, possessed a normal T cell compartment, pointing to developmental defects as a cause of peripheral T cell lymphopenia in PPARδmut mice. These findings implicate PPAR-δ as a regulator of the metabolic program during thymocyte and T cell growth.
Collapse
Affiliation(s)
- Fei Linda Zhao
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jeeyoon Jennifer Ahn
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Edward L Y Chen
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tae Joon Yi
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada
| | | | - David Spaner
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and
| | - Shannon E Dunn
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; .,Toronto General Hospital Research Institute, Toronto, Ontario M5G 2C4, Canada.,Women's College Health Research Institute, Toronto, Ontario M5G 1N8, Canada
| |
Collapse
|
40
|
Crystal structure of the ferric homotetrameric β 4 human hemoglobin. Biophys Chem 2018; 240:9-14. [DOI: 10.1016/j.bpc.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 11/21/2022]
|
41
|
Yang X, Zhang H, Shang J, Liu G, Xia T, Zhao C, Sun G, Dou H. Comparative analysis of the blood transcriptomes between wolves and dogs. Anim Genet 2018; 49:291-302. [PMID: 29953636 DOI: 10.1111/age.12675] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2018] [Indexed: 12/24/2022]
Abstract
Dogs were domesticated by human and originated from wolves. Their evolutionary relationships have attracted much scientific interest due to their genetic affinity but different habitats. To identify the differences between dogs and wolves associated with domestication, we analysed the blood transcriptomes of wolves and dogs by RNA-Seq. We obtained a total of 30.87 Gb of raw reads from two dogs and three wolves using RNA-Seq technology. Comparisons of the wolf and dog transcriptomes revealed 524 genes differentially expressed genes between them. We found that some genes related to immune function (DCK, ICAM4, GAPDH and BSG) and aerobic capacity (HBA1, HBA2 and HBB) were more highly expressed in the wolf. Six differentially expressed genes related to the innate immune response (CCL23, TRIM10, DUSP10, RAB27A, CLEC5A and GCH1) were found in the wolf by a Gene Ontology enrichment analysis. Immune system development was also enriched only in the wolf group. The ALAS2, HMBS and FECH genes, shown to be enriched by the Kyoto Encyclopedia of Genes and Genomes analysis, were associated with the higher aerobic capacity and hypoxia endurance of the wolf. The results suggest that the wolf might have greater resistance to pathogens, hypoxia endurance and aerobic capacity than dogs do.
Collapse
Affiliation(s)
- X Yang
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - H Zhang
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - J Shang
- College of Information Science and Engineering, Qufu Normal University, Yantai North Road No. 80, Rizhao, Shandong, 276826, China
| | - G Liu
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - T Xia
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - C Zhao
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - G Sun
- College of Life Science, Qufu Normal University, Jingxuan West Road No. 57, Qufu, Shandong, 273165, China
| | - H Dou
- Dailake National Nature Reserve, Manzhouli Road No. 16, Hulunbuir, Inner Mongolia, 021000, China
| |
Collapse
|
42
|
Li P, Du L, Li W, Fan Z, Zeng D, Chen H, Zhou L, Yi Y, Yang N, Dou K, Yue B, Li J. Generation and characterization of the blood transcriptome of Macaca thibetana and comparative analysis with M. mulatta. MOLECULAR BIOSYSTEMS 2018; 13:1121-1130. [PMID: 28428989 DOI: 10.1039/c6mb00771f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transcriptome profiles provide a large transcript sequence data set for genomic study, particularly in organisms that have no accurate genome data published. The Tibetan macaque (Macaca thibetana) is commonly considered to be an endemic species to China and an important animal in biomedical research in the present day. In the present study, we report the de novo assembly and characterization of the blood transcriptome of the Tibetan macaque from three individuals, and we also sequenced the blood transcriptome of the rhesus macaque (Macaca mulatta) for comparison. Using RNA-seq technology, 138 million sequencing reads of the M. thibetana transcriptome were generated. The assembly included 327 871 transcripts with an N50 of 1571 bp. According to the sequence similarity search, 80 317 (24.5%) transcripts were annotated in the nr protein database. All transcripts from M. thibetana and M. mulatta were functionally classified and compared using GO and KEGG analyses. The two transcriptomes were different in the GO term of nutrient reservoir activity, and in the KEGG subcategories of signaling molecules and interaction, infectious diseases, cell growth and death, and immune system. The transcriptomes in this study would provide a valuable resource for future functional and comparative genomic studies, and even for biological studies of this non-human primate.
Collapse
Affiliation(s)
- Peng Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, Sichuan 610064, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hsp90 chaperones hemoglobin maturation in erythroid and nonerythroid cells. Proc Natl Acad Sci U S A 2018; 115:E1117-E1126. [PMID: 29358373 DOI: 10.1073/pnas.1717993115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Maturation of adult (α2β2) and fetal hemoglobin (α2γ2) tetramers requires that heme be incorporated into each globin. While hemoglobin alpha (Hb-α) relies on a specific erythroid chaperone (alpha Hb-stabilizing protein, AHSP), the other chaperones that may help mature the partner globins (Hb-γ or Hb-β) in erythroid cells, or may enable nonerythroid cells to express mature Hb, are unknown. We investigated a role for heat-shock protein 90 (hsp90) in Hb maturation in erythroid precursor cells that naturally express Hb-α with either Hb-γ (K562 and HiDEP-1 cells) or Hb-β (HUDEP-2) and in nonerythroid cell lines that either endogenously express Hb-αβ (RAW and A549) or that we transfected to express the globins. We found the following: (i) AHSP and hsp90 associate with distinct globin partners in their immature heme-free states (AHSP with apo-Hbα, and hsp90 with apo-Hbβ or Hb-γ) and that hsp90 does not associate with mature Hb. (ii) Hsp90 stabilizes the apo-globins and helps to drive their heme insertion reactions, as judged by pharmacologic hsp90 inhibition or by coexpression of an ATP-ase defective hsp90. (iii) In nonerythroid cells, heme insertion into all globins became hsp90-dependent, which may explain how mixed Hb tetramers can mature in cells that do not express AHSP. Together, our findings uncover a process in which hsp90 first binds to immature, heme-free Hb-γ or Hb-β, drives their heme insertion process, and then dissociates to allow their heterotetramer formation with Hb-α. Thus, in driving heme insertion, hsp90 works in concert with AHSP to generate functional Hb tetramers during erythropoiesis.
Collapse
|
44
|
Meimaridou E, Goldsworthy M, Chortis V, Fragouli E, Foster PA, Arlt W, Cox R, Metherell LA. NNT is a key regulator of adrenal redox homeostasis and steroidogenesis in male mice. J Endocrinol 2018; 236:13-28. [PMID: 29046340 PMCID: PMC5744559 DOI: 10.1530/joe-16-0638] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/18/2017] [Indexed: 01/10/2023]
Abstract
Nicotinamide nucleotide transhydrogenase, NNT, is a ubiquitous protein of the inner mitochondrial membrane with a key role in mitochondrial redox balance. NNT produces high concentrations of NADPH for detoxification of reactive oxygen species by glutathione and thioredoxin pathways. In humans, NNT dysfunction leads to an adrenal-specific disorder, glucocorticoid deficiency. Certain substrains of C57BL/6 mice contain a spontaneously occurring inactivating Nnt mutation and display glucocorticoid deficiency along with glucose intolerance and reduced insulin secretion. To understand the underlying mechanism(s) behind the glucocorticoid deficiency, we performed comprehensive RNA-seq on adrenals from wild-type (C57BL/6N), mutant (C57BL/6J) and BAC transgenic mice overexpressing Nnt (C57BL/6JBAC). The following results were obtained. Our data suggest that Nnt deletion (or overexpression) reduces adrenal steroidogenic output by decreasing the expression of crucial, mitochondrial antioxidant (Prdx3 and Txnrd2) and steroidogenic (Cyp11a1) enzymes. Pathway analysis also revealed upregulation of heat shock protein machinery and haemoglobins possibly in response to the oxidative stress initiated by NNT ablation. In conclusion, using transcriptomic profiling in adrenals from three mouse models, we showed that disturbances in adrenal redox homeostasis are mediated not only by under expression of NNT but also by its overexpression. Further, we demonstrated that both under expression or overexpression of NNT reduced corticosterone output implying a central role for it in the control of steroidogenesis. This is likely due to a reduction in the expression of a key steroidogenic enzyme, Cyp11a1, which mirrored the reduction in corticosterone output.
Collapse
Affiliation(s)
- E Meimaridou
- Centre for EndocrinologyWilliam Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, London, UK
| | - M Goldsworthy
- MRC Harwell InstituteGenetics of Type 2 Diabetes, Mammalian Genetics Unit, Oxfordshire, UK
| | - V Chortis
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - E Fragouli
- Centre for EndocrinologyWilliam Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, London, UK
| | - P A Foster
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - W Arlt
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - R Cox
- MRC Harwell InstituteGenetics of Type 2 Diabetes, Mammalian Genetics Unit, Oxfordshire, UK
| | - L A Metherell
- Centre for EndocrinologyWilliam Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, London, UK
| |
Collapse
|
45
|
Karhu T, Akiyama K, Vuolteenaho O, Bergmann U, Naito T, Tatemoto K, Herzig KH. Isolation of new ligands for orphan receptor MRGPRX1-hemorphins LVV-H7 and VV-H7. Peptides 2017; 96:61-66. [PMID: 28867075 DOI: 10.1016/j.peptides.2017.08.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/15/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022]
Abstract
The human MAS-related G protein-coupled receptor X1 (MRGPRX1) is a member of the GPCR family. The receptor is primate specific and expressed in the sensory neurons of dorsal root ganglion and trigeminal ganglion, where it is considered to be involved in the pain perception. The MRGPRX1 has unusual binding mechanism, as it is activated by several different ligands as well as several different fragments of precursor proteins. Thus, we hypothesize that it is activated by several unknown compounds as well since the receptor is still classified as orphan. Here, we describe the isolation of two novel endogenous ligands for the MRGPRX1 from human platelet preparation. The isolated ligands are hemoglobin β-chain fragments, known members of the hemorphin family.
Collapse
Affiliation(s)
- T Karhu
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - K Akiyama
- Okinawa Institute of Science and Technology Graduate University, Tancha, Onna-son, Okinawa, Japan
| | - O Vuolteenaho
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Nordlab Oulu, Oulu, Finland
| | - U Bergmann
- Biocenter Oulu, University of Oulu, Oulu, Finland; Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - T Naito
- Okinawa Institute of Science and Technology Graduate University, Tancha, Onna-son, Okinawa, Japan; Research Institute of Natural-Drug Leads, Kanagawa University, Hiratsuka, Kanagawa, Japan
| | - K Tatemoto
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - K-H Herzig
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland; Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
46
|
Connor DE, Chaitanya GV, Chittiboina P, McCarthy P, Scott LK, Schrott L, Minagar A, Nanda A, Alexander JS. Variations in the cerebrospinal fluid proteome following traumatic brain injury and subarachnoid hemorrhage. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2017; 24:169-183. [PMID: 28549769 PMCID: PMC7303909 DOI: 10.1016/j.pathophys.2017.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Proteomic analysis of cerebrospinal fluid (CSF) has shown great promise in identifying potential markers of injury in neurodegenerative diseases [1-13]. Here we compared CSF proteomes in healthy individuals, with patients diagnosed with traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) in order to characterize molecular biomarkers which might identify these different clinical states and describe different molecular mechanisms active in each disease state. METHODS Patients presenting to the Neurosurgery service at the Louisiana State University Hospital-Shreveport with an admitting diagnosis of TBI or SAH were prospectively enrolled. Patients undergoing CSF sampling for diagnostic procedures were also enrolled as controls. CSF aliquots were subjected to 2-dimensional gel electrophoresis (2D GE) and spot percentage densities analyzed. Increased or decreased spot expression (compared to controls) was defined in terms of in spot percentages, with spots showing consistent expression change across TBI or SAH specimens being followed up by Matrix-Assisted Laser Desorption/Ionization mass spectrometry (MALDI-MS). Polypeptide masses generated were matched to known standards using a search of the NCBI and/or GenPept databases for protein matches. Eight hundred fifteen separately identifiable polypeptide migration spots were identified on 2D GE gels. MALDI-MS successfully identified 13 of 22 selected 2D GE spots as recognizable polypeptides. RESULTS Statistically significant changes were noted in the expression of fibrinogen, carbonic anhydrase-I (CA-I), peroxiredoxin-2 (Prx-2), both α and β chains of hemoglobin, serotransferrin (Tf) and N-terminal haptoglobin (Hp) in TBI and SAH specimens, as compared to controls. The greatest mean fold change among all specimens was seen in CA-I and Hp at 30.7 and -25.7, respectively. TBI specimens trended toward greater mean increases in CA-I and Prx-2 and greater mean decreases in Hp and Tf. CONCLUSIONS Consistent CSF elevation of CA-I and Prx-2 with concurrent depletion of Hp and Tf may represent a useful combination of biomarkers for the prediction of severity and prognosis following brain injury.
Collapse
Affiliation(s)
- David E Connor
- Baptist Health Neurosurgery Arkansas, Little Rock, AR, United States.
| | - Ganta V Chaitanya
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD, United States.
| | - Paul McCarthy
- Department of Medicine, Sect. of Nephrology, University of Maryland, Baltimore, MD, United States.
| | - L Keith Scott
- Department of Critical Care Medicine, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Lisa Schrott
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Anil Nanda
- Department of Neurosurgery, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| |
Collapse
|
47
|
Non-conventional role of haemoglobin beta in breast malignancy. Br J Cancer 2017; 117:994-1006. [PMID: 28772282 PMCID: PMC5625664 DOI: 10.1038/bjc.2017.247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 05/31/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Besides its role as oxygen transporter, recent findings suggest that haemoglobin beta (HBB) may have roles in other contexts. Methods: We evaluated the impact of HBB expression in primary human breast cancers, and in breast cancer cell lines overexpressing HBB by in vitro and in vivo studies. Publicly available microarray databases were used to perform multivariate survival analyses. Results: A significantly higher expression of HBB was observed in invasive carcinoma histotypes vs in situ counterparts, along with a positive correlation between HBB and the Ki67 proliferation marker. HBB-overexpressing breast cancer cells migrate and invade more, show HIF-1α upregulation and their conditioned media enhances angiogenesis. Blocking the oxygen-binding site of HBB reverts the increase of migration and HIF-1α upregulation observed in HBB-overexpressing breast cancer cells. Orthotopically implanted MDA-MB-231 overexpressing HBB (MDA-HBB) generated tumours with faster growth rate and increased neoangiogenesis. Moreover, local recurrence and visceral metastases were observed only in MDA-HBB-implanted mice. Similar results were observed with 4T1 mouse breast cancer cells. Finally, bioinformatics analyses of public data sets correlated high HBB expression with lower overall survival. Conclusions: HBB expression increases breast cancer cells aggressiveness and associates with poor prognosis, pointing to HBB as a novel biomarker for breast cancer progression.
Collapse
|
48
|
Zheng Y, Miyamoto DT, Wittner BS, Sullivan JP, Aceto N, Jordan NV, Yu M, Karabacak NM, Comaills V, Morris R, Desai R, Desai N, Emmons E, Milner JD, Lee RJ, Wu CL, Sequist LV, Haas W, Ting DT, Toner M, Ramaswamy S, Maheswaran S, Haber DA. Expression of β-globin by cancer cells promotes cell survival during blood-borne dissemination. Nat Commun 2017; 8:14344. [PMID: 28181495 PMCID: PMC5321792 DOI: 10.1038/ncomms14344] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
Metastasis-competent circulating tumour cells (CTCs) experience oxidative stress in the bloodstream, but their survival mechanisms are not well defined. Here, comparing single-cell RNA-Seq profiles of CTCs from breast, prostate and lung cancers, we observe consistent induction of β-globin (HBB), but not its partner α-globin (HBA). The tumour-specific origin of HBB is confirmed by sequence polymorphisms within human xenograft-derived CTCs in mouse models. Increased intracellular reactive oxygen species (ROS) in cultured breast CTCs triggers HBB induction, mediated through the transcriptional regulator KLF4. Depletion of HBB in CTC-derived cultures has minimal effects on primary tumour growth, but it greatly increases apoptosis following ROS exposure, and dramatically reduces CTC-derived lung metastases. These effects are reversed by the anti-oxidant N-Acetyl Cysteine. Conversely, overexpression of HBB is sufficient to suppress intracellular ROS within CTCs. Altogether, these observations suggest that β-globin is selectively deregulated in cancer cells, mediating a cytoprotective effect during blood-borne metastasis. Circulating tumour cells contribute to metastatic spread. Here the authors find that beta-chain of haemoglobin is overexpressed in those cells and protects them from oxidative stress, prolonging their survival in circulation and thereby increasing the likelihood of metastasis formation.
Collapse
Affiliation(s)
- Yu Zheng
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - David T Miyamoto
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Ben S Wittner
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - James P Sullivan
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Nicola Aceto
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Nicole Vincent Jordan
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Min Yu
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Nezihi Murat Karabacak
- Center for Bioengineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Valentine Comaills
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Robert Morris
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Rushil Desai
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Niyati Desai
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Erin Emmons
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - John D Milner
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Richard J Lee
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Chin-Lee Wu
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Lecia V Sequist
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Wilhelm Haas
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - David T Ting
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Mehmet Toner
- Center for Bioengineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Sridhar Ramaswamy
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Shyamala Maheswaran
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Daniel A Haber
- Massachusetts General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA.,Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
49
|
Saha D, Koli S, Patgaonkar M, Reddy KVR. Expression of hemoglobin-α and β subunits in human vaginal epithelial cells and their functional significance. PLoS One 2017; 12:e0171084. [PMID: 28178273 PMCID: PMC5298339 DOI: 10.1371/journal.pone.0171084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/16/2017] [Indexed: 11/18/2022] Open
Abstract
Hemoglobin (Hb) is a major protein involved in transport of oxygen (O2). It consists of Hb-α and Hb-β subunits, which are normally expressed by cells of erythroid lineage. However, till recently, it was not known whether non-erythroid cells like vaginal cells synthesize Hb and whether it has any functional significance. Therefore, we designed the following objectives: (1) to establish in-vitro culture system of human primary vaginal epithelial cells (hPVECs), (2) to determine whether Hb-α and Hb-β proteins are truly synthesized by hPVECs, (3) to evaluate the effect of LPS (lipopolysaccharide) on the expression of Hb-α and Hb-β proteins (4) to decipher the significance of the Hb-α and Hb-β expression in hPVECs and (5) to determine the molecular mechanism regulating the expression of Hb-α in hPVECs. To accomplish these studies, we applied a battery of assays such as RT-PCR, qRT-PCR, Flow cytometry, western blot, and immunofluorescence, Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). The results revealed the expression of Hb-α and Hb-β at both mRNA and protein level in hPVECs. The expression was significantly upregulated following LPS treatment (10μg/ml for 6 hrs) and these results are comparable with the expression induced by LPS in human vaginal epithelial cell line (VK2/E6E7). These cells constitutively produced low levels of pro-inflammatory (IL-6) and anti-inflammatory (IL-10) cytokines. Also, the response of phosphorylated (p65)-NF-κB to LPS was upregulated with increased expression of IL-6, Toll-like receptor-4 (TLR4) and human beta defensin-1 (hBD-1) in hPVECs and VK2/E6E7 cells. However, Bay 11-7082 treatment (5μM for 24 hrs) could neutralize the effect of LPS-induced p65-NF-κB activity and represses the production`of Hb-α and Hb-β. The results of EMSA revealed the presence of putative binding sites of NF-κB in the human Hb-α promoter region (nt-115 to -106). ChIP analysis confirmed the binding of NF-κB to Hb-α promoter. In conclusion, the present findings revealed for the first time that hPVECs synthesized Hb-α and Hb-β and the expression is comparable with the expression of VK2/E6E7 cells. The identification of NF-κB regulatory sequences in Hb-α promoter, whose activation is associated with immune response of hPVECs, indicating Hb-α and Hb-β may act as an endogenous antimicrobial defense protein against vaginal inflammation/infections.
Collapse
Affiliation(s)
- Debarchana Saha
- Division of Molecular Immunology and Microbiology (MIM), National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Swanand Koli
- Division of Molecular Immunology and Microbiology (MIM), National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Mandar Patgaonkar
- Division of Molecular Immunology and Microbiology (MIM), National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Kudumula Venkata Rami Reddy
- Division of Molecular Immunology and Microbiology (MIM), National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
- * E-mail:
| |
Collapse
|
50
|
Garton T, Keep RF, Hua Y, Xi G. Brain iron overload following intracranial haemorrhage. Stroke Vasc Neurol 2016; 1:172-184. [PMID: 28959481 PMCID: PMC5435218 DOI: 10.1136/svn-2016-000042] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 12/15/2022] Open
Abstract
Intracranial haemorrhages, including intracerebral haemorrhage (ICH), intraventricular haemorrhage (IVH) and subarachnoid haemorrhage (SAH), are leading causes of morbidity and mortality worldwide. In addition, haemorrhage contributes to tissue damage in traumatic brain injury (TBI). To date, efforts to treat the long-term consequences of cerebral haemorrhage have been unsatisfactory. Incident rates and mortality have not showed significant improvement in recent years. In terms of secondary damage following haemorrhage, it is becoming increasingly apparent that blood components are of integral importance, with haemoglobin-derived iron playing a major role. However, the damage caused by iron is complex and varied, and therefore, increased investigation into the mechanisms by which iron causes brain injury is required. As ICH, IVH, SAH and TBI are related, this review will discuss the role of iron in each, so that similarities in injury pathologies can be more easily identified. It summarises important components of normal brain iron homeostasis and analyses the existing evidence on iron-related brain injury mechanisms. It further discusses treatment options of particular promise.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|