1
|
Tozatto‐Maio K, Rós FA, Weinlich R, Rocha V. Inflammatory pathways and anti-inflammatory therapies in sickle cell disease. Hemasphere 2024; 8:e70032. [PMID: 39698332 PMCID: PMC11655128 DOI: 10.1002/hem3.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 07/12/2024] [Accepted: 09/17/2024] [Indexed: 12/20/2024] Open
Abstract
Sickle cell disease (SCD) is a monogenic disease, resulting from a single-point mutation, that presents a complex pathophysiology and high clinical heterogeneity. Inflammation stands as a prominent characteristic of SCD. Over the past few decades, the role of different cells and molecules in the regulation of the inflammatory process has been elucidated. In conjunction with the polymerization of hemoglobin S (HbS), intravascular hemolysis, which releases free heme, HbS, and hemoglobin-related damage-associated molecular patterns, initiates multiple inflammatory pathways that are not yet fully comprehended. These complex phenomena lead to a vicious cycle that perpetuates vaso-occlusion, hemolysis, and inflammation. To date, few inflammatory biomarkers can predict disease complications; conversely, there is a plethora of therapies that reduce inflammation in SCD, although clinical outcomes vary widely. Importantly, whether the clinical heterogeneity and complications are related to the degree of inflammation is not known. This review aims to further our understanding of the roles of main immune cells, and other inflammatory factors, as potential prognostic biomarkers for predicting clinical outcomes or identifying novel treatments for SCD.
Collapse
Affiliation(s)
- Karina Tozatto‐Maio
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Felipe A. Rós
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Ricardo Weinlich
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
| | - Vanderson Rocha
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
- Instituto D'Or de Ensino e Pesquisa, Rede D'OrSao PauloBrazil
- Department of Hematology, Churchill HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Rarick KR, Li K, Teng RJ, Jing X, Martin DP, Xu H, Jones DW, Hogg N, Hillery CA, Garcia G, Day BW, Naylor S, Pritchard KA. Sterile inflammation induces vasculopathy and chronic lung injury in murine sickle cell disease. Free Radic Biol Med 2024; 215:112-126. [PMID: 38336101 PMCID: PMC11290318 DOI: 10.1016/j.freeradbiomed.2024.01.052] [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: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Murine sickle cell disease (SCD) results in damage to multiple organs, likely mediated first by vasculopathy. While the mechanisms inducing vascular damage remain to be determined, nitric oxide bioavailability and sterile inflammation are both considered to play major roles in vasculopathy. Here, we investigate the effects of high mobility group box-1 (HMGB1), a pro-inflammatory damage-associated molecular pattern (DAMP) molecule on endothelial-dependent vasodilation and lung morphometrics, a structural index of damage in sickle (SS) mice. SS mice were treated with either phosphate-buffered saline (PBS), hE-HMGB1-BP, an hE dual-domain peptide that binds and removes HMGB1 from the circulation via the liver, 1-[4-(aminocarbonyl)-2-methylphenyl]-5-[4-(1H-imidazol-1-yl)phenyl]-1H-pyrrole-2-propanoic acid (N6022) or N-acetyl-lysyltyrosylcysteine amide (KYC) for three weeks. Human umbilical vein endothelial cells (HUVEC) were treated with recombinant HMGB1 (r-HMGB1), which increases S-nitrosoglutathione reductase (GSNOR) expression by ∼80%, demonstrating a direct effect of HMGB1 to increase GSNOR. Treatment of SS mice with hE-HMGB1-BP reduced plasma HMGB1 in SS mice to control levels and reduced GSNOR expression in facialis arteries isolated from SS mice by ∼20%. These changes were associated with improved endothelial-dependent vasodilation. Treatment of SS mice with N6022 also improved vasodilation in SS mice suggesting that targeting GSNOR also improves vasodilation. SCD decreased protein nitrosothiols (SNOs) and radial alveolar counts (RAC) and increased GSNOR expression and mean linear intercepts (MLI) in lungs from SS mice. The marked changes in pulmonary morphometrics and GSNOR expression throughout the lung parenchyma in SS mice were improved by treating with either hE-HMGB1-BP or KYC. These data demonstrate that murine SCD induces vasculopathy and chronic lung disease by an HMGB1- and GSNOR-dependent mechanism and suggest that HMGB1 and GSNOR might be effective therapeutic targets for reducing vasculopathy and chronic lung disease in humans with SCD.
Collapse
Affiliation(s)
- Kevin R Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA
| | - Keguo Li
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ru-Jeng Teng
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA
| | - Xigang Jing
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Dustin P Martin
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hao Xu
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Deron W Jones
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Cheryl A Hillery
- Department of Pediatrics, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Department of Pediatrics, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Guilherme Garcia
- Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | | | - Kirkwood A Pritchard
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; ReNeuroGen LLC, Milwaukee, WI, 53122, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
3
|
Silva M, Faustino P. From Stress to Sick(le) and Back Again-Oxidative/Antioxidant Mechanisms, Genetic Modulation, and Cerebrovascular Disease in Children with Sickle Cell Anemia. Antioxidants (Basel) 2023; 12:1977. [PMID: 38001830 PMCID: PMC10669666 DOI: 10.3390/antiox12111977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Sickle cell anemia (SCA) is a genetic disease caused by the homozygosity of the HBB:c.20A>T mutation, which results in the production of hemoglobin S (HbS). In hypoxic conditions, HbS suffers autoxidation and polymerizes inside red blood cells, altering their morphology into a sickle shape, with increased rigidity and fragility. This triggers complex pathophysiological mechanisms, including inflammation, cell adhesion, oxidative stress, and vaso-occlusion, along with metabolic alterations and endocrine complications. SCA is phenotypically heterogeneous due to the modulation of both environmental and genetic factors. Pediatric cerebrovascular disease (CVD), namely ischemic stroke and silent cerebral infarctions, is one of the most impactful manifestations. In this review, we highlight the role of oxidative stress in the pathophysiology of pediatric CVD. Since oxidative stress is an interdependent mechanism in vasculopathy, occurring alongside (or as result of) endothelial dysfunction, cell adhesion, inflammation, chronic hemolysis, ischemia-reperfusion injury, and vaso-occlusion, a brief overview of the main mechanisms involved is included. Moreover, the genetic modulation of CVD in SCA is discussed. The knowledge of the intricate network of altered mechanisms in SCA, and how it is affected by different genetic factors, is fundamental for the identification of potential therapeutic targets, drug development, and patient-specific treatment alternatives.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
- Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
4
|
Hebbel RP, Milbauer L, Wei P. A novel promoter of endothelial dysfunction in African Americans: Relevance to sickle cell anaemia. Br J Haematol 2023; 203:e71-e73. [PMID: 37485546 DOI: 10.1111/bjh.18993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Affiliation(s)
- Robert P Hebbel
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Liming Milbauer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Peng Wei
- Department of Biostatistics, University of Texas MD Anderson Center, Houston, Texas, USA
| |
Collapse
|
5
|
Gbotosho OT, Gollamudi J, Hyacinth HI. The Role of Inflammation in The Cellular and Molecular Mechanisms of Cardiopulmonary Complications of Sickle Cell Disease. Biomolecules 2023; 13:381. [PMID: 36830749 PMCID: PMC9953727 DOI: 10.3390/biom13020381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Cardiopulmonary complications remain the major cause of mortality despite newer therapies and improvements in the lifespan of patients with sickle cell disease (SCD). Inflammation has been identified as a major risk modifier in the pathogenesis of SCD-associated cardiopulmonary complications in recent mechanistic and observational studies. In this review, we discuss recent cellular and molecular mechanisms of cardiopulmonary complications in SCD and summarize the most recent evidence from clinical and laboratory studies. We emphasize the role of inflammation in the onset and progression of these complications to better understand the underlying pathobiological processes. We also discuss future basic and translational research in addressing questions about the complex role of inflammation in the development of SCD cardiopulmonary complications, which may lead to promising therapies and reduce morbidity and mortality in this vulnerable population.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| | - Jahnavi Gollamudi
- Division of Hematology & Oncology, Department of Internal Medicine, 3125 Eden Avenue, ML 0562, Cincinnati, OH 45219-0562, USA
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| |
Collapse
|
6
|
Marchesani S, Bertaina V, Marini O, Cossutta M, Di Mauro M, Rotulo GA, Palma P, Sabatini L, Petrone MI, Frati G, Monteleone G, Palumbo G, Ceglie G. Inflammatory status in pediatric sickle cell disease: Unravelling the role of immune cell subsets. Front Mol Biosci 2023; 9:1075686. [PMID: 36703915 PMCID: PMC9871358 DOI: 10.3389/fmolb.2022.1075686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction: The mutation of the beta-globin gene that causes sickle cell disease (SCD) results in pleiotropic effects, such as hemolysis and vaso-occlusive crisis that can induce inflammatory mechanisms with deleterious consequences on the organism. Moreover, SCD patients display an increased susceptibility to infections. Few studies are currently available that evaluate a wide immunological profile in a pediatric population. This study proposes an evaluation of the immune profile in subjects with SCD in a pediatric population through a detailed analysis by flow cytometry. Methods and Materials: Peripheral blood samples from 53 pediatric patients with SCD (mean age 9.8 years, interquartile range 9 years) were obtained and then analyzed by flow cytometry, in order to evaluate changes in the immune populations compared to 40 healthy donors (mean age 7.3 years, interquartile range 9.5 years). Results: Our data showed an increase in neutrophils (with a reduction in the CD62L + subpopulation) and monocytes (with a decrease in HLA-DRlow monocytes) with normal values of lymphocytes in SCD patients. In the lymphocyte subpopulations analysis we observed lower values of CD4+ T cells (with higher number of memory and central memory T lymphocytes) with increased frequency of CD8+ T cells (with a predominant naive pattern). Moreover, we observed higher values of CD39+ Tregs and lower HLA-DR+ and CD39- T cells with an increased Th17, Th1-17 and Th2 response. Conclusion: We observed immunological alterations typical of an inflammatory status (increase in activated neutrophils and monocytes) associated with a peculiar Treg pattern (probably linked to a body attempt to minimize inflammation intrinsic to SCD). Furthermore, we highlighted a T helper pathway associated with inflammation in line with other studies. Our data showed that immunological markers may have an important role in the understanding the pathophysiology of SCD and in optimizing targeted therapeutic strategies for each patient.
Collapse
Affiliation(s)
- Silvio Marchesani
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,*Correspondence: Silvio Marchesani,
| | - Valentina Bertaina
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Olivia Marini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Women’s and Children’s Health Department, Hematology-Oncology Clinic and Laboratory, University of Padova, Padova, Italy
| | - Matilde Cossutta
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Margherita Di Mauro
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Gioacchino Andrea Rotulo
- Clinical and Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children Hospital, IRCCS, Rome, Italy,Department of Neuroscience, Rehabilitation Ophthalmology Genetics Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Paolo Palma
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,Clinical and Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Letizia Sabatini
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Maria Isabella Petrone
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Giacomo Frati
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giulia Monteleone
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Palumbo
- University Department of Pediatrics, Bambino Gesù Children’s Hospital, University of Rome Tor Vergata, Rome, Italy,Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giulia Ceglie
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
7
|
Kour D, Ali M, Khajuria P, Sharma K, Ghosh P, Kaur S, Mahajan S, Ramajayan P, Bharate SS, Bhardwaj S, Sawant SD, Reddy DS, Kumar A. Flurbiprofen inhibits heme induced NLRP3 inflammasome in Berkeley sickle cell disease mice. Front Pharmacol 2023; 14:1123734. [PMID: 37180702 PMCID: PMC10171431 DOI: 10.3389/fphar.2023.1123734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Sickle cell disease (SCD) is accompanied by several complications, which emanate from the sickling of erythrocytes due to a point mutation in the β-globin chain of hemoglobin. Sickled erythrocytes are unable to move smoothly through small blood capillaries and therefore, cause vaso occlusion and severe pain. Apart from pain, continuous lysis of fragile sickled erythrocytes leads to the release of heme, which is a strong activator of the NLRP3 inflammasome, thus producing chronic inflammation in sickle cell disease. In this study, we identified flurbiprofen among other COX-2 inhibitors to be a potent inhibitor of heme-induced NLRP3 inflammasome. We found that apart from being a nociceptive agent, flurbiprofen exerts a strong anti-inflammatory effect by suppressing NF-κB signaling, which was evidenced by reduced levels of TNF-α and IL-6 in wild-type and sickle cell disease Berkeley mice models. Our data further demonstrated the protective effect of flurbiprofen on liver, lungs, and spleen in Berkeley mice. The current sickle cell disease pain management regime relies mainly on opiate drugs, which is accompanied by several side effects without modifying the sickle cell disease-related pathology. Considering the potent role of flurbiprofen in inhibiting NLRP3 inflammasome and other inflammatory cytokines in sickle cell disease, our data suggests that it can be explored further for better sickle cell disease pain management along with the possibility of disease modification.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Mehboob Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Palash Ghosh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sukhleen Kaur
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Surbhi Mahajan
- Department of Pathology, Government Medical College, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Sonali S. Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai, India
| | - Subhash Bhardwaj
- Department of Pathology, Government Medical College, Jammu, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - D. Srinivasa Reddy
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| |
Collapse
|
8
|
Arafat AMA, Botros SKA, Afifi R, Zayed SA, Fateen M. Serum Level of High-Mobility Group Box Protein 1 as a Potential Treatment Target in Egyptian Sickle Cell Disease Patients. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background:
During tissue injury, high mobility group box 1 (HMGB1) is passively released from necrotic cells and actively secreted by inflammatory cells. Extracellular HMGB1 acts as an amplifier of Toll-Like Receptor (TLR)-dependent inflammation rather than a primary trigger of inflammation. We studied HMGB1 quantitative trait locus reference sequence 2249825 (rs2249825) and its serum level in both sickle cell disease (SCD) patients and healthy subjects to explore its possible role in the pathogenesis of vaso-occlusive crises (VOCs).
Methods:
HMGB1 rs2249825 was assayed in peripheral blood samples using real-time polymerase chain reaction (RT-PCR). While the serum level was assayed using a two-site enzyme-linked immunosorbent technique (ELISA).
Results:
Both the SCD patients and the control group had comparable HMGB1 rs2249825 genotype frequencies (P-value >0.05). SCD patients at their steady-state showed statistically significantly higher serum HMGB1 levels than the healthy controls, a median of 0.6 ng/ml with a range of 0.1- 85 ng/ml versus a median of 0.3 ng/ml and a range of 0.1-3 ng/ml (P-value <0.001), respectively. Statistically significant skewed high serum HMGB1 in the VOC samples in contrast to the steady-state samples was observed in the SCD patients with a median of 3.2 ng/ml and a range of 0.3-76.4 ng/ml versus a median of 0.2 ng/ml and a range 0.2-7.4 ng/ml (P-value <0.0001), respectively.
Conclusion:
HMGB1 could have a role in the VOC pathogenesis, hence it is suggested as a potential additive therapeutic target in SCD in general and in vaso-occlusions in specific.
Keywords:
Sickle cell disease, HMGB1, Hemoglobin S
Collapse
|
9
|
Folic Acid Protects Melanocytes from Oxidative Stress via Activation of Nrf2 and Inhibition of HMGB1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1608586. [PMID: 34917229 PMCID: PMC8670940 DOI: 10.1155/2021/1608586] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 12/21/2022]
Abstract
Vitiligo is a cutaneous depigmentation disease due to loss of epidermal melanocytes. Accumulating evidence has indicated that oxidative stress plays a vital role in vitiligo via directly destructing melanocytes and triggering inflammatory response that ultimately undermines melanocytes. Folic acid (FA), an oxidized form of folate with high bioavailability, exhibits potent antioxidant properties and shows therapeutic potential in multiple oxidative stress-related diseases. However, whether FA safeguards melanocytes from oxidative damages remains unknown. In this study, we first found that FA relieved melanocytes from H2O2-induced abnormal growth and apoptosis. Furthermore, FA enhanced the activity of antioxidative enzymes and remarkably reduced intracellular ROS levels in melanocytes. Subsequently, FA effectively activated nuclear factor E2-related factor 2 (Nrf2) pathway, and Nrf2 knockdown blocked the protective effects of FA on H2O2-treated melanocytes. Additionally, FA inhibited the production of proinflammatory HMGB1 in melanocytes under oxidative stress. Taken together, our findings support the protective effects of FA on human melanocytes against oxidative injury via the activation of Nrf2 and the inhibition of HMGB1, thus indicating FA as a potential therapeutic agent for the treatment of vitiligo.
Collapse
|
10
|
Liu H, Chu S, Wu Z. Loss of toll-like receptor 4 ameliorates cardiovascular dysfunction in aged mice. Immun Ageing 2021; 18:42. [PMID: 34740366 PMCID: PMC8569991 DOI: 10.1186/s12979-021-00251-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022]
Abstract
Background Toll-like receptor 4 (TLR4) is a pattern recognition receptor of the innate immune system. TLR4 contributes to many aging-related chronic diseases. However, whether TLR4 is involved in cardiovascular injury during the aging process has not been investigated. Methods The effects of TLR4 on the cardiovascular system of aged mice were investigated in TLR4−/− mice. An intraperitoneal glucose tolerance test (IPGTT) and insulin sensitivity test (IST) were conducted to evaluate global insulin sensitivity. Echocardiography was used to measure cardiac structure and performance. An isolated artery ring assay was used to measure the vasodilator function of the thoracic aorta. The inflammatory response was reflected by the serum concentration of cytokines. Results TLR4 expression increased in the hearts and aortas of mice in an age-dependent manner. Loss of TLR4 increased insulin sensitivity in aged mice. Moreover, loss of TLR4 improved cardiac performance and endothelium-dependent vascular relaxation in aged mice. Importantly, the increases in serum inflammatory cytokines and oxidative stress in the heart and aorta were also inhibited by TLR4 deficiency. Conclusion In summary, loss of TLR4 improved cardiac performance and endothelium-dependent vascular relaxation in aged mice. The reduced inflammatory responses and oxidative stress may be the reason for the protective effects of TLR4 deficiency during aging. Our study indicates that targeting TLR4 is a potential therapeutic strategy for preventing aging-related cardiovascular disease.
Collapse
Affiliation(s)
- Huan Liu
- Anesthesiology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Shujuan Chu
- Anesthesiology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| | - Zhilin Wu
- Anesthesiology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
11
|
Hebbel RP, Vercellotti GM. Multiple inducers of endothelial NOS (eNOS) dysfunction in sickle cell disease. Am J Hematol 2021; 96:1505-1517. [PMID: 34331722 PMCID: PMC9292023 DOI: 10.1002/ajh.26308] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/26/2022]
Abstract
A characteristic aspect of the robust, systemic inflammatory state in sickle cell disease is dysfunction of endothelial nitric oxide synthase (eNOS). We identify 10 aberrant endothelial cell inputs, present in the specific sickle context, that are known to have the ability to cause eNOS dysfunction. These are: endothelial arginase depletion, asymmetric dimethylarginine, complement activation, endothelial glycocalyx degradation, free fatty acids, inflammatory mediators, microparticles, oxidized low density lipoproteins, reactive oxygen species, and Toll‐like receptor 4 signaling ligands. The effect of true eNOS dysfunction on clinical testing using flow‐mediated dilation can be simulated by two known examples of endothelial dysfunction mimicry (hemoglobin consumption of NO; and oxidation of smooth muscle cell soluble guanylate cyclase). This lends ambiguity to interpretation of such clinical testing. The presence of these multiple perturbing factors argues that a therapeutic approach targeting only a single injurious endothelial input (or either example of mimicry) would not be sufficiently efficacious. This would seem to argue for identifying therapeutics that directly protect eNOS function or application of multiple therapeutic approaches.
Collapse
Affiliation(s)
- Robert P. Hebbel
- Division of Hematology‐Oncology‐Transplantation, Department of Medicine University of Minnesota Medical School Minneapolis Minnesota USA
| | - Gregory M. Vercellotti
- Division of Hematology‐Oncology‐Transplantation, Department of Medicine University of Minnesota Medical School Minneapolis Minnesota USA
| |
Collapse
|
12
|
Wang Q, Zennadi R. The Role of RBC Oxidative Stress in Sickle Cell Disease: From the Molecular Basis to Pathologic Implications. Antioxidants (Basel) 2021; 10:antiox10101608. [PMID: 34679742 PMCID: PMC8533084 DOI: 10.3390/antiox10101608] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 01/14/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited monogenic disorder and the most common severe hemoglobinopathy in the world. SCD is characterized by a point mutation in the β-globin gene, which results in hemoglobin (Hb) S production, leading to a variety of mechanistic and phenotypic changes within the sickle red blood cell (RBC). In SCD, the sickle RBCs are the root cause of the disease and they are a primary source of oxidative stress since sickle RBC redox state is compromised due to an imbalance between prooxidants and antioxidants. This imbalance in redox state is a result of a continuous production of reactive oxygen species (ROS) within the sickle RBC caused by the constant endogenous Hb autoxidation and NADPH oxidase activation, as well as by a deficiency in the antioxidant defense system. Accumulation of non-neutralized ROS within the sickle RBCs affects RBC membrane structure and function, leading to membrane integrity deficiency, low deformability, phosphatidylserine exposure, and release of micro-vesicles. These oxidative stress-associated RBC phenotypic modifications consequently evoke a myriad of physiological changes involved in multi-system manifestations. Thus, RBC oxidative stress in SCD can ultimately instigate major processes involved in organ damage. The critical role of the sickle RBC ROS production and its regulation in SCD pathophysiology are discussed here.
Collapse
|
13
|
Pitanga TN, Santana SS, Zanette DL, Guarda CC, Santiago RP, Maffili VV, Lima JB, Carvalho GQ, Filho JR, Ferreira JRD, Aleluia MM, Nascimento VML, Carvalho MOS, Lyra IM, Borges VM, Oliveira RR, Goncalves MS. Effect of lysed and non-lysed sickle red cells on the activation of NLRP3 inflammasome and LTB4 production by mononuclear cells. Inflamm Res 2021; 70:823-834. [PMID: 34196737 DOI: 10.1007/s00011-021-01461-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE AND DESIGN This study tested the hypothesis that sickle red blood cell (SS-RBC) can induce inflammasome NLRP3 components gene expression in peripheral blood mononuclear cells (PBMCs) as well as interleukin-1β (IL-1β) and leukotriene B4 (LTB4) production. Additionally, we investigated the effect of hydroxyurea (HU) treatment in these inflammatory markers. METHODS PBMCs from healthy donors (AA-PBMC) were challenged with intact and lysed RBCs from SCA patients (SS-RBC) and from healthy volunteers (AA-RBC). NLRP3, IL-1β, IL-18 and Caspase-1 gene expression levels were assessed by quantitative PCR (qPCR). IL-1β protein levels and LTB4 were measured by ELISA. RESULTS We observed that lysed SS-RBC induced the expression of inflammasome NLRP3 components, but this increase was more prominent for CASP1 and IL18 expression levels. Moreover, we observed that intact SS-RBC induced higher production of IL-1β and LTB4 than lysed SS-RBC. Although SCA patients treated with HU have a reduction in NLRP3 gene expression and LTB4 production, this treatment did not modulate the expression of other inflammasome components or IL-1β production. CONCLUSIONS Thus, our data suggest that caspase-1, IL-1β and IL-18 may contribute to the inflammatory status observed in SCA and that HU treatment may not interfere in this inflammatory pathway.
Collapse
Affiliation(s)
- Thassila N Pitanga
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil.,Universidade Católica do Salvador (UCSAL), Salvador, Bahia, Brazil
| | - Sânzio S Santana
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil.,Universidade Católica do Salvador (UCSAL), Salvador, Bahia, Brazil
| | - Dalila L Zanette
- Fundação Oswaldo Cruz, Instituto Carlos Chagas (ICC-FIOCRUZ/PR), Curitiba, Paraná, Brazil
| | - Caroline C Guarda
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Rayra P Santiago
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Vitor V Maffili
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Jonilson B Lima
- Universidade Federal do Oeste da Bahia (UFOB), Barreiras, Bahia, Brazil
| | - Graziele Q Carvalho
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Jaime R Filho
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | | | - Milena M Aleluia
- Universidade Estadual de Santa Cruz (UESC), Ilhéus, Bahia, Brazil
| | - Valma M L Nascimento
- Fundação de Hematologia e Hemoterapia da Bahia (HEMOBA), Salvador, Bahia, Brazil
| | - Magda O S Carvalho
- Hospital Universitário Professor Edgard Santos (HUPES), UFBA, Salvador, Bahia, Brazil
| | - Isa M Lyra
- Hospital Universitário Professor Edgard Santos (HUPES), UFBA, Salvador, Bahia, Brazil
| | - Valéria M Borges
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Ricardo R Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Marilda S Goncalves
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil. .,Universidade Federal da Bahia (UFBA), Salvador, Bahia, Brazil.
| |
Collapse
|
14
|
|
15
|
Nader E, Conran N, Romana M, Connes P. Vasculopathy in Sickle Cell Disease: From Red Blood Cell Sickling to Vascular Dysfunction. Compr Physiol 2021; 11:1785-1803. [PMID: 33792905 DOI: 10.1002/cphy.c200024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sickle cell disease (SCD) is a hereditary disorder that leads to the production of an abnormal hemoglobin, hemoglobin S (HbS). HbS polymerizes in deoxygenated conditions, which can prompt red blood cell (RBC) sickling and leaves the RBCs more rigid, fragile, and prone to hemolysis. SCD patients suffer from a plethora of complications, ranging from acute complications, such as characteristic, frequent, and debilitating vaso-occlusive episodes to chronic organ damage. While RBC sickling is the primary event at the origin of vaso-occlusive processes, other factors that can further increase RBC transit times in the microcirculation may also be required to precipitate vaso-occlusive processes. The adhesion of RBC and leukocytes to activated endothelium and the formation of heterocellular aggregates, as well as increased blood viscosity, are among the mechanisms involved in slowing the progress of RBCs in deoxygenated vascular areas, favoring RBC sickling and promoting vascular occlusion. Chronic inflammatory processes and oxidative stress, which are perpetuated by hemolytic events and ischemia-reperfusion injury, result in this pan cellular activation and some acute events, such as stroke and acute chest syndrome, as well as chronic end-organ damage. Furthermore, impaired vasodilation and vasomotor hyperresponsiveness in SCD also contribute to vaso-occlusive processes. Treating SCD as a vascular disease in addition to its hematological perspective, the present article looks at the interplay between abnormal RBC physiology/integrity, vascular dysfunction and clinical severity in SCD, and discusses existing therapies and novel drugs in development that may ameliorate vascular complications in the disease. © 2021 American Physiological Society. Compr Physiol 11:1785-1803, 2021.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| |
Collapse
|
16
|
Teng RJ, Jing X, Martin DP, Hogg N, Haefke A, Konduri GG, Day BW, Naylor S, Pritchard KA. N-acetyl-lysyltyrosylcysteine amide, a novel systems pharmacology agent, reduces bronchopulmonary dysplasia in hyperoxic neonatal rat pups. Free Radic Biol Med 2021; 166:73-89. [PMID: 33607217 PMCID: PMC8009865 DOI: 10.1016/j.freeradbiomed.2021.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 01/26/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is caused primarily by oxidative stress and inflammation. To induce BPD, neonatal rat pups were raised in hyperoxic (>90% O2) environments from day one (P1) until day ten (P10) and treated with N-acetyl-lysyltyrosylcysteine amide (KYC). In vivo studies showed that KYC improved lung complexity, reduced myeloperoxidase (MPO) positive (+) myeloid cell counts, MPO protein, chlorotyrosine formation, increased endothelial cell CD31 expression, decreased 8-OH-dG and Cox-1/Cox-2, HMGB1, RAGE, TLR4, increased weight gain and improved survival in hyperoxic pups. EPR studies confirmed that MPO reaction mixtures oxidized KYC to a KYC thiyl radical. Adding recombinant HMGB1 to the MPO reaction mixture containing KYC resulted in KYC thiylation of HMGB1. In rat lung microvascular endothelial cell (RLMVEC) cultures, KYC thiylation of RLMVEC proteins was increased the most in RLMVEC cultures treated with MPO + H2O2, followed by H2O2, and then KYC alone. KYC treatment of hyperoxic pups decreased total HMGB1 in lung lysates, increased KYC thiylation of HMGB1, terminal HMGB1 thiol oxidation, decreased HMGB1 association with TLR4 and RAGE, and shifted HMGB1 in lung lysates from a non-acetylated to a lysyl-acetylated isoform, suggesting that KYC reduced lung cell death and that recruited immune cells had become the primary source of HMGB1 released into the hyperoxic lungs. MPO-dependent and independent KYC-thiylation of Keap1 were both increased in RLMVEC cultures. Treating hyperoxic pups with KYC increased KYC thiylation and S-glutathionylation of Keap1, and Nrf2 activation. These data suggest that KYC is a novel system pharmacological agent that exploits MPO to inhibit toxic oxidant production and is oxidized into a thiyl radical that inactivates HMGB1, activates Nrf2, and increases antioxidant enzyme expression to improve lung complexity and reduce BPD in hyperoxic rat pups.
Collapse
Affiliation(s)
- Ru-Jeng Teng
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Xigang Jing
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Dustin P Martin
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA; ReNeuroGen LLC, Milwaukee, WI, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aaron Haefke
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Girija G Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, WI, USA
| | | | | | - Kirkwood A Pritchard
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA; ReNeuroGen LLC, Milwaukee, WI, USA.
| |
Collapse
|
17
|
Zhang P, Nguyen J, Abdulla F, Nelson AT, Beckman JD, Vercellotti GM, Belcher JD. Soluble MD-2 and Heme in Sickle Cell Disease Plasma Promote Pro-Inflammatory Signaling in Endothelial Cells. Front Immunol 2021; 12:632709. [PMID: 33841413 PMCID: PMC8033004 DOI: 10.3389/fimmu.2021.632709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/10/2021] [Indexed: 12/22/2022] Open
Abstract
Recent evidence indicates that hemolysis in sickle cell disease (SCD) promotes inflammation via innate immune signaling through toll-like receptor 4 (TLR4). Free heme released by hemolyzed red blood cells can bind to myeloid differentiation factor-2 (MD-2) and activate TLR4 pro-inflammatory signaling on endothelium to promote vaso-occlusion and acute chest syndrome in murine models of SCD. MD-2 is co-expressed with TLR4 on cell membranes, but in inflammatory conditions, soluble MD-2 (sMD-2) is elevated in plasma. sMD-2 levels were significantly increased in human and murine sickle (SS) plasma as compared to normal (AA) plasma. Human umbilical vein endothelial cells (HUVEC) and human lung microvascular endothelial cells incubated with human SS plasma had significant increases in pro-inflammatory IL-8, IL-6, and soluble VCAM-1 secretion compared to endothelial cells incubated with AA plasma. The increase in HUVEC IL-8 secretion was blocked by depletion of sMD-2 from SS plasma and enhanced by the addition of sMD-2 to AA plasma. The TLR4 signaling inhibitor, TAK-242, inhibited HUVEC IL-8 secretion in response to SS plasma by 85%. Heme-agarose pull-down assays and UV/Vis spectroscopy demonstrated that heme binds to sMD-2. Hemopexin, a high affinity heme-binding protein, inhibited HUVEC IL-8 secretion induced by SS plasma or SS and AA plasma supplemented with sMD-2. These data suggest that sMD-2 bound to heme might play an important role in pro-inflammatory signaling by endothelium in SCD.
Collapse
Affiliation(s)
- Ping Zhang
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Julia Nguyen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Fuad Abdulla
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Alexander T Nelson
- University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Joan D Beckman
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - John D Belcher
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
18
|
Silva-Junior AL, Garcia NP, Cardoso EC, Dias S, Tarragô AM, Fraiji NA, Gomes MS, Amaral LR, Teixeira-Carvalho A, Martins-Filho OA, De Paula EV, Costa AG, Malheiro A. Immunological Hallmarks of Inflammatory Status in Vaso-Occlusive Crisis of Sickle Cell Anemia Patients. Front Immunol 2021; 12:559925. [PMID: 33776989 PMCID: PMC7990896 DOI: 10.3389/fimmu.2021.559925] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Sickle Cell Anemia (SCA) is the most common genetic disorder around the world. The mutation in the β-globin gene is responsible for a higher hemolysis rate, with further involvement of immunological molecules, especially cytokines, chemokines, growth factors, and anaphylatoxins. These molecules are responsible for inducing and attracting immune cells into circulation, thus contributing to increases in leukocytes and other pro-inflammatory mediators, and can culminate in a vaso-occlusive crisis (VOC). This study aimed to characterize the levels of these molecules in SCA patients in different clinical conditions in order to identify potential hallmarks of inflammation in these patients. An analytical prospective study was conducted using the serum of SCA patients in steady-state (StSt; n = 27) and VOC (n = 22), along with 53 healthy donors (HD). Samples from the VOC group were obtained on admission and on discharge, in the convalescent phase (CV). Levels of chemokines (CXCL8, CXCL10, CL2, CLL3, CCL4, CL5, and CCL11), cytokines (IL-1β, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12p70, IL-13, IL-17A, TNF-α, and IFN-γ) and growth factors (VEGF, FGFb, PDGF-BB, GM-CSF, and G-CSF) were measured using a Luminex assay, and anaphylatoxins (C3a, C4a, and C5a) were measured using Cytometric Bead Array. SCA patients in StSt showed a pro-inflammatory profile, and were indicated as being higher producers of CCL2, IL-1β, IL-12p70, IFN-γ, IL-17A, and GM-CSF, while VOC is highlighted by molecules IL-4 and IL-5, but also IL-2, IL-7, PDGF-BB, and G-CSF. PDGF-BB and IL-1ra seemed to be two important hallmarks for the acute-to-chronic stage, due to their significant decrease after crisis inflammation and statistical difference in VOC and CV groups. These molecules show higher levels and a strong correlation with other molecules in VOC. Furthermore, they remain at higher levels even after crisis recovery, which suggest their importance in the role of inflammation during crisis and participation in immune cell adhesion and activation. These results support a relevant role of cytokines, neutrophil and monocytes, since these may act as markers of VOC inflammation in SCA patients.
Collapse
Affiliation(s)
- Alexander Leonardo Silva-Junior
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Nadja Pinto Garcia
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Evilázio Cunha Cardoso
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Stephanny Dias
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Andrea Monteiro Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Nelson Abrahim Fraiji
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Matheus Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Laurence Rodrigues Amaral
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Andréa Teixeira-Carvalho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Erich Vinicius De Paula
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Escola de Ciências Médicas, Universidade de Campinas, Campinas, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
- Escola de Enfermagem de Manaus, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| |
Collapse
|
19
|
Li B, Peng X, Li H, Chen F, Chen Y, Zhang Y, Le K. The performance of the alarmin HMGB1 in pediatric diseases: From lab to clinic. Immun Inflamm Dis 2021; 9:8-30. [PMID: 33140586 PMCID: PMC7860603 DOI: 10.1002/iid3.370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The ubiquitously expressed nonhistone nuclear protein high-mobility group box protein 1 (HMGB1) has different functions related to posttranslational modifications and cellular localization. In the nucleus, HMGB1 modulates gene transcription, replication and DNA repair as well as determines chromosomal architecture. When the post-transcriptional modified HMGB1 is released into the extracellular space, it triggers several physiological and pathological responses and initiates innate immunity through interacting with its reciprocal receptors (i.e., TLR4/2 and RAGE). The effect of HMGB1-mediated inflammatory activation on different systems has received increasing attention. HMGB1 is now considered to be an alarmin and participates in multiple inflammation-related diseases. In addition, HMGB1 also affects the occurrence and progression of tumors. However, most studies involving HMGB1 have been focused on adults or mature animals. Due to differences in disease characteristics between children and adults, it is necessary to clarify the role of HMGB1 in pediatric diseases. METHODS AND RESULTS Through systematic database retrieval, this review aimed to first elaborate the characteristics of HMGB1 under physiological and pathological conditions and then discuss the clinical significance of HMGB1 in the pediatric diseases according to different systems. CONCLUSIONS HMGB1 plays an important role in a variety of pediatric diseases and may be used as a diagnostic biomarker and therapeutic target for new strategies for the prevention and treatment of pediatric diseases.
Collapse
Affiliation(s)
- Bo Li
- Department of CardiologyChildren's Hospital of Hebei Province Affiliated to Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xin Peng
- Department of OtolaryngologyThe Affiliated Children's Hospital of Nanchang UniversityNanchangJiangxiChina
| | - He Li
- Department of Urology SurgeryQilu Children's Hospital of Shandong UniversityJinanShandongChina
| | - Fei Chen
- Department of Child Health CareQilu Children's Hospital of Shandong UniversityJinanShandongChina
| | - Yuxia Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, and Rehabilitation Centre, Children's HospitalChongqing Medical UniversityChongqingYuzhongChina
| | - Yingqian Zhang
- Department of CardiologyChildren's Hospital of Hebei Province Affiliated to Hebei Medical UniversityShijiazhuangHebeiChina
| | - Kai Le
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| |
Collapse
|
20
|
A Unique Monocyte Transcriptome Discriminates Sickle Cell Disease From Other Hereditary Hemolytic Anemias and Shows the Particular Importance of Lipid and Interferon Signaling. Hemasphere 2021; 5:e531. [PMID: 33604514 PMCID: PMC7886403 DOI: 10.1097/hs9.0000000000000531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 12/17/2020] [Indexed: 01/17/2023] Open
|
21
|
Khaibullina A, Almeida LEF, Kamimura S, Zerfas PM, Smith ML, Vogel S, Wakim P, Vasconcelos OM, Quezado MM, Horkayne-Szakaly I, Quezado ZMN. Sickle cell disease mice have cerebral oxidative stress and vascular and white matter abnormalities. Blood Cells Mol Dis 2021; 86:102493. [PMID: 32927249 PMCID: PMC7686096 DOI: 10.1016/j.bcmd.2020.102493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Strokes are feared complications of sickle cell disease (SCD) and yield significant neurologic and neurocognitive deficits. However, even without detectable strokes, SCD patients have significant neurocognitive deficits in domains of learning and memory, processing speed and executive function. In these cases, mechanisms unrelated to major cerebrovascular abnormalities likely underlie these deficits. While oxidative stress and stress-related signaling pathways play a role in SCD pathophysiology, their role in cerebral injury remains unknown. We have shown that Townes and BERK SCD mice, while not having strokes, recapitulate neurocognitive deficits reported in humans. We hypothesized that cognitive deficits in SCD mice are associated with cerebral oxidative stress. We showed that SCD mice have increased levels of reactive oxygen species, protein carbonylation, and lipid peroxidation in hippocampus and cortex, thus suggesting increased cerebral oxidative stress. Further, cerebral oxidative stress was associated with caspase-3 activity alterations and vascular endothelial abnormalities, white matter changes, and disruption of the blood brain barrier, similar to those reported after ischemic/oxidative injury. Additionally, after repeated hypoxia/reoxygenation exposure, homozygous Townes had enhanced microglia activation. Our findings indicate that oxidative stress and stress-induced tissue damage is increased in susceptible brain regions, which may, in turn, contribute to neurocognitive deficits in SCD mice.
Collapse
Affiliation(s)
- Alfia Khaibullina
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Patricia M Zerfas
- Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Meghann L Smith
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Sebastian Vogel
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Paul Wakim
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Olavo M Vasconcelos
- Neuromuscular Clinic, Electromyography Laboratory, Intraoperative Neurophysiology Monitoring Sections, Veterans Health Administration Medical Center, Virginia Commonwealth University, Richmond, VA 23249, United States of America
| | - Martha M Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Iren Horkayne-Szakaly
- Neuropathology and Ophthalmic Pathology, Joint Pathology Center, Defense Health Agency, Silver Spring, MD 20910, United States of America
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States of America.
| |
Collapse
|
22
|
Gbotosho OT, Kapetanaki MG, Kato GJ. The Worst Things in Life are Free: The Role of Free Heme in Sickle Cell Disease. Front Immunol 2021; 11:561917. [PMID: 33584641 PMCID: PMC7873693 DOI: 10.3389/fimmu.2020.561917] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hemolysis is a pathological feature of several diseases of diverse etiology such as hereditary anemias, malaria, and sepsis. A major complication of hemolysis involves the release of large quantities of hemoglobin into the blood circulation and the subsequent generation of harmful metabolites like labile heme. Protective mechanisms like haptoglobin-hemoglobin and hemopexin-heme binding, and heme oxygenase-1 enzymatic degradation of heme limit the toxicity of the hemolysis-related molecules. The capacity of these protective systems is exceeded in hemolytic diseases, resulting in high residual levels of hemolysis products in the circulation, which pose a great oxidative and proinflammatory risk. Sickle cell disease (SCD) features a prominent hemolytic anemia which impacts the phenotypic variability and disease severity. Not only is circulating heme a potent oxidative molecule, but it can act as an erythrocytic danger-associated molecular pattern (eDAMP) molecule which contributes to a proinflammatory state, promoting sickle complications such as vaso-occlusion and acute lung injury. Exposure to extracellular heme in SCD can also augment the expression of placental growth factor (PlGF) and interleukin-6 (IL-6), with important consequences to enthothelin-1 (ET-1) secretion and pulmonary hypertension, and potentially the development of renal and cardiac dysfunction. This review focuses on heme-induced mechanisms that are implicated in disease pathways, mainly in SCD. A special emphasis is given to heme-induced PlGF and IL-6 related mechanisms and their role in SCD disease progression.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maria G. Kapetanaki
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gregory J. Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
23
|
Hebbel RP, Belcher JD, Vercellotti GM. The multifaceted role of ischemia/reperfusion in sickle cell anemia. J Clin Invest 2020; 130:1062-1072. [PMID: 32118586 DOI: 10.1172/jci133639] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sickle cell anemia is a unique disease dominated by hemolytic anemia and vaso-occlusive events. The latter trigger a version of ischemia/reperfusion (I/R) pathobiology that is singular in its origin, cyclicity, complexity, instability, perpetuity, and breadth of clinical consequences. Specific clinical features are probably attributable to local I/R injury (e.g., stroke syndromes) or remote organ injury (e.g., acute chest syndrome) or the systematization of inflammation (e.g., multifocal arteriopathy). Indeed, by fashioning an underlying template of endothelial dysfunction and vulnerability, the robust inflammatory systematization no doubt contributes to all sickle pathology. In this Review, we highlight I/R-targeting therapeutics shown to improve microvascular blood flow in sickle transgenic mice undergoing I/R, and we suggest how such insights might be translated into human therapeutic strategies.
Collapse
|
24
|
Shet AS, Lizarralde-Iragorri MA, Naik RP. The molecular basis for the prothrombotic state in sickle cell disease. Haematologica 2020; 105:2368-2379. [PMID: 33054077 PMCID: PMC7556662 DOI: 10.3324/haematol.2019.239350] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The genetic and molecular basis of sickle cell disease (SCD) has long since been characterized but the pathophysiological basis is not entirely defined. How a red cell hemolytic disorder initiates inflammation, endothelial dysfunction, coagulation activation and eventually leads to vascular thrombosis, is yet to be elucidated. Recent evidence has demonstrated a high frequency of unprovoked/recurrent venous thromboembolism (VTE) in SCD, with an increased risk of mortality among patients with a history of VTE. Here, we thoroughly review the molecular basis for the prothrombotic state in SCD, specifically highlighting emerging evidence for activation of overlapping inflammation and coagulation pathways, that predispose to venous thromboembolism. We share perspectives in managing venous thrombosis in SCD, highlighting innovative therapies with the potential to influence the clinical course of disease and reduce thrombotic risk, while maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Arun S. Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda
| | | | - Rakhi P. Naik
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
25
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
26
|
Lizarralde-Iragorri MA, Shet AS. Sickle Cell Disease: A Paradigm for Venous Thrombosis Pathophysiology. Int J Mol Sci 2020; 21:ijms21155279. [PMID: 32722421 PMCID: PMC7432404 DOI: 10.3390/ijms21155279] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is an important cause of vascular morbidity and mortality. Many risk factors have been identified for venous thrombosis that lead to alterations in blood flow, activate the vascular endothelium, and increase the propensity for blood coagulation. However, the precise molecular and cellular mechanisms that cause blood clots in the venous vasculature have not been fully elucidated. Patients with sickle cell disease (SCD) demonstrate all the risk factors for venous stasis, activated endothelium, and blood hypercoagulability, making them particularly vulnerable to VTE. In this review, we will discuss how mouse models have elucidated the complex vascular pathobiology of SCD. We review the dysregulated pathways of inflammation and coagulation in SCD and how the resultant hypercoagulable state can potentiate thrombosis through down-regulation of vascular anticoagulants. Studies of VTE pathogenesis using SCD mouse models may provide insight into the intersection between the cellular and molecular processes involving inflammation and coagulation and help to identify novel mechanistic pathways.
Collapse
|
27
|
Garcia NP, Júnior ALS, Soares GAS, Costa TCC, dos Santos APC, Costa AG, Tarragô AM, Martins RN, do Carmo Leão Pontes F, de Almeida EG, de Paula EV, Martins-Filho OA, Malheiro A. Sickle Cell Anemia Patients Display an Intricate Cellular and Serum Biomarker Network Highlighted by TCD4+CD69+ Lymphocytes, IL-17/MIP-1 β, IL-12/VEGF, and IL-10/IP-10 Axis. J Immunol Res 2020; 2020:4585704. [PMID: 32411797 PMCID: PMC7199620 DOI: 10.1155/2020/4585704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/14/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sickle cell anemia (SCA) is associated with a chronic proinflammatory state characterized by elevated leukocyte count, mortality from severe recurrent infections, and subsequent vasoocclusive complications with leukocyte adhesion to the endothelium and increased plasma levels of inflammatory cytokines. The immune system has a close connection with morbidity in SCA, but further studies are needed to uncover the involvement of innate and adaptive immunities in modulating the SCA physiopathology. We performed measurements of the frequency of innate and adaptive immunity cells, cytokines, chemokines, and growth factors and immunophenotyping of Toll-like receptor and adhesion molecule expression in the blood of SCA patients and healthy donors to evaluate the different profiles of these biomarkers, the relationship among them, and their correlation to laboratory records and death risk. Material and Methods. Immunophenotyping of cells, Toll-like receptors, and adhesion molecules were performed from peripheral blood samples of SCA patients and healthy donors by flow cytometry and cytokine/chemokine/growth factor measurement by the Luminex technique performed from the serum of the same subjects. RESULTS Cells of adaptive immunity such as IL-12, IL-17, and IL-10 cytokines; IL-8, IP-10, MIP-1α, MIP-1β, and RANTES chemokines; and VEGF, FGF-basic, and GM-CSF growth factors were higher in SCA patients than healthy donors regardless of any laboratorial and clinical condition. However, high death risk appears to have relevant biomarkers. CONCLUSION In the SCA pathophysiology at steady state, there is a broad immunological biomarker crosstalk highlighted by TCD4+CD69+ lymphocytes, IL-12 and IL-17 inflammatory and IL-10 regulatory cytokines, MIP-1α, MIP-1β, and IP-10 chemokines, and VEGF growth factor. High expression of TLR2 in monocytes and VLA-4 in TCD8+ lymphocytes and high levels of MIP-1β and RANTES appear to be relevant in high death risk conditions. The high reticulocytosis and high death risk conditions present common correlations, and there seems to be a balance by the Th2 profile.
Collapse
Affiliation(s)
- Nadja Pinto Garcia
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), 69077-000 Manaus, AM, Brazil
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Alexander Leonardo S. Júnior
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
| | - Geyse Adriana S. Soares
- Programa de Apoio a Iniciação Científica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Thainá Cristina C. Costa
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
| | - Alicia Patrine C. dos Santos
- Programa de Apoio a Iniciação Científica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), 69077-000 Manaus, AM, Brazil
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
| | - Andréa Monteiro Tarragô
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Rejane Nina Martins
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Flávia do Carmo Leão Pontes
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
| | - Emerson Garcia de Almeida
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
| | - Erich Vinícius de Paula
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
- Departamento de Clínica Médica da Faculdade de Ciências Médicas da UNICAMP, 13083-970 Campinas, SP, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou/Fiocruz Minas, 30190-002 Belo Horizonte, MG, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), 69077-000 Manaus, AM, Brazil
- Laboratório de Genômica, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), 69050-001 Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas a Hematologia, Universidade Estadual do Amazonas (PPCAH/UEA), 69065-001 Manaus, AM, Brazil
| |
Collapse
|
28
|
The platelet NLRP3 inflammasome is upregulated in sickle cell disease via HMGB1/TLR4 and Bruton tyrosine kinase. Blood Adv 2019; 2:2672-2680. [PMID: 30333099 DOI: 10.1182/bloodadvances.2018021709] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/17/2018] [Indexed: 01/01/2023] Open
Abstract
A key inflammatory mechanism recently identified in platelets involves the Nod-like receptor nucleotide-binding domain leucine-rich repeat containing protein 3 (NLRP3) and Bruton tyrosine kinase (BTK), which control activation of caspase-1 within inflammasome complexes. We investigated platelet caspase-1 activity in the context of sickle cell disease (SCD) directly in platelets isolated from SCD patients (n = 24) and indirectly by incubating platelets from healthy subjects with plasma obtained from SCD patients (n = 20), both in steady state and during an acute pain crisis (paired samples). The platelet NLRP3 inflammasome was upregulated in SCD patients under steady state conditions compared with healthy controls, and it was further upregulated when patients experienced an acute pain crisis. The results were consistent with indirect platelet assays, in which SCD plasma increased caspase-1 activity of platelets from healthy subjects in an NLRP3-dependent fashion. The damage-associated molecular pattern molecule high-mobility group box 1 (HMGB1) was elevated in plasma of SCD subjects compared with healthy controls and correlated with caspase-1 activity in platelets. Pharmacological or antibody-mediated inhibition of HMGB1, Toll-like receptor 4, and BTK interfered with sickle plasma-induced platelet caspase-1 activation. In Townes SCD mice, caspase-1 activity and aggregation of circulating platelets were elevated, which was suppressed by IV injection of an NLRP3 inhibitor and the BTK inhibitor ibrutinib. Activation of the platelet NLRP3 inflammasome in SCD may have diagnostic and therapeutic implications.
Collapse
|
29
|
Nolfi-Donegan D, Pradhan-Sundd T, Pritchard KA, Hillery CA. Redox Signaling in Sickle Cell Disease. CURRENT OPINION IN PHYSIOLOGY 2019; 9:26-33. [PMID: 31240269 DOI: 10.1016/j.cophys.2019.04.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sickle cell disease (SCD) is characterized by chronic hemolysis and repeated episodes of vascular occlusion leading to progressive organ injury. SCD is characterized by unbalanced, simultaneous pro-oxidant and anti-oxidant processes at the molecular, cellular and tissue levels, with the majority of reactions tipped in favor of pro-oxidant pathways. In this brief review we discuss new findings regarding how oxidized hemin, hemolysis, mitochondrial dysfunction and the innate immune system generate oxidative stress while hemopexin, haptoglobin, heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) may provide protection in human and murine SCD. We will also describe recent clinical trials showing beneficial effects of antioxidant therapy in SCD.
Collapse
Affiliation(s)
- Deirdre Nolfi-Donegan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tirthadipa Pradhan-Sundd
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kirkwood A Pritchard
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Cheryl A Hillery
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Vercellotti GM, Dalmasso AP, Schaid TR, Nguyen J, Chen C, Ericson ME, Abdulla F, Killeen T, Lindorfer MA, Taylor RP, Belcher JD. Critical role of C5a in sickle cell disease. Am J Hematol 2019; 94:327-337. [PMID: 30569594 DOI: 10.1002/ajh.25384] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/16/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022]
Abstract
Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.
Collapse
Affiliation(s)
- Gregory M. Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | | | - Terry R. Schaid
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Chunsheng Chen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Marna E. Ericson
- Department of Dermatology; University of Minnesota; Minneapolis Minnesota
| | - Fuad Abdulla
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Trevor Killeen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Margaret A. Lindorfer
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - John D. Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
31
|
Tran H, Mittal A, Sagi V, Luk K, Nguyen A, Gupta M, Nguyen J, Lamarre Y, Lei J, Guedes A, Gupta K. Mast Cells Induce Blood Brain Barrier Damage in SCD by Causing Endoplasmic Reticulum Stress in the Endothelium. Front Cell Neurosci 2019; 13:56. [PMID: 30837844 PMCID: PMC6389721 DOI: 10.3389/fncel.2019.00056] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
Endothelial dysfunction underlies the pathobiology of cerebrovascular disease. Mast cells are located in close proximity to the vasculature, and vasoactive mediators released upon their activation can promote endothelial activation leading to blood brain barrier (BBB) dysfunction. We examined the mechanism of mast cell-induced endothelial activation via endoplasmic reticulum (ER) stress mediated P-selectin expression in a transgenic mouse model of sickle cell disease (SCD), which shows BBB dysfunction. We used mouse brain endothelial cells (mBECs) and mast cells-derived from skin of control and sickle mice to examine the mechanisms involved. Compared to control mouse mast cell conditioned medium (MCCM), mBECs incubated with sickle mouse MCCM showed increased, structural disorganization and swelling of the ER and Golgi, aggregation of ribosomes, ER stress marker proteins, accumulation of galactose-1-phosphate uridyl transferase, mitochondrial dysfunction, reactive oxygen species (ROS) production, P-selectin expression and mBEC permeability. These effects of sickle-MCCM on mBEC were inhibited by Salubrinal, a reducer of ER stress. Histamine levels in the plasma, skin releasate and in mast cells of sickle mice were higher compared to control mice. Compared to control BBB permeability was increased in sickle mice. Treatment of mice with imatinib, Salubrinal, or P-selectin blocking antibody reduced BBB permeability in sickle mice. Mast cells induce endothelial dysfunction via ER stress-mediated P-selectin expression. Mast cell activation contributes to ER stress mediated endothelial P-selectin expression leading to increased endothelial permeability and impairment of BBB. Targeting mast cells and/or ER stress has the potential to ameliorate endothelial dysfunction in SCD and other pathobiologies.
Collapse
Affiliation(s)
- Huy Tran
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Aditya Mittal
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Varun Sagi
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Kathryn Luk
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Aithanh Nguyen
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Mihir Gupta
- Department of Neurosurgery, University of California, San Diego, San Diego, CA, United States
| | - Julia Nguyen
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Yann Lamarre
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Jianxun Lei
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Alonso Guedes
- Anesthesia and Pain Medicine, Veterinary Clinical Science Department, College of Veterinary Medicine, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Kalpna Gupta
- Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, Medical School, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
32
|
Safwat NA, Kenny MA. Soluble receptor for advanced glycation end products as a vasculopathy biomarker in sickle cell disease. Pediatr Res 2018; 84:869-874. [PMID: 30367158 DOI: 10.1038/s41390-018-0221-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/01/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Soluble forms of RAGE (sRAGE) have been found circulating in plasma and tissues. Evidence is accruing in human subjects linking levels of sRAGE to oxidative stress in many disorders. Because sickle cell disease (SCD) is a state of oxidative stress, we tested the hypothesis that circulating sRAGE levels may be involved in the vascular pathology of SCD. OBJECTIVES To determine the sRAGE levels in children and adolescents with SCD and investigate their association with markers of hemolysis, iron overload, and SCD-related organ complications. SUBJECTS AND METHODS The level of sRAGE was measured in 40 children and adolescent with SCD compared with 40 healthy controls using enzyme-linked immunosorbent assay (ELISA). RESULTS sRAGE was significantly higher in patients compared with controls (p < 0.001) and was elevated in patients with history of stroke, acute lung syndrome, and frequency of sickling crisis or serum ferritin > 2500 (p < 0.05). Patients with high sRAGE levels are candidates for chelation. sRAGE was positively correlated with HbS% (r = 0.422, p = 0.007), LDH (r = 0.329, p = 0.038), and serum ferritin levels (r = 0.516, p = 0.001). Multivariable regression analysis proved that both HbS% and serum ferritin were significant independent factors affecting sRAGE level (p < 0.05). CONCLUSION Our findings suggest that sRAGE may be considered as a marker for vascular dysfunction in SCD patients.
Collapse
Affiliation(s)
- Nesma Ahmed Safwat
- Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | |
Collapse
|
33
|
Kalpatthi R, Novelli EM. Measuring success: utility of biomarkers in sickle cell disease clinical trials and care. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:482-492. [PMID: 30504349 PMCID: PMC6246014 DOI: 10.1182/asheducation-2018.1.482] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Progress in the care of sickle cell disease (SCD) has been hampered by the extreme complexity of the SCD phenotype despite its monogenic inheritance. While epidemiological studies have identified clinical biomarkers of disease severity, with a few exceptions, these have not been routinely incorporated in clinical care algorithms. Furthermore, existing biomarkers have been poorly apt at providing objective parameters to diagnose sickle cell crisis, the hallmark, acute complication of SCD. The repercussions of these diagnostic limitations are reflected in suboptimal care and scarcity of adequate outcome measures for clinical research. Recent progress in molecular and imaging diagnostics has heralded a new era of personalized medicine in SCD. Precision medicine strategies are particularly timely, since molecular therapeutics are finally on the horizon. This chapter will summarize the existing evidence and promising data on biomarkers for clinical care and research in SCD.
Collapse
Affiliation(s)
- Ram Kalpatthi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA; and
| | - Enrico M. Novelli
- Division of Hematology/Oncology and UPMC Heart, Lung and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
34
|
Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:263-292. [PMID: 30332562 DOI: 10.1146/annurev-pathmechdis-012418-012838] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the discovery of sickle cell disease (SCD) in 1910, enormous strides have been made in the elucidation of the pathogenesis of its protean complications, which has inspired recent advances in targeted molecular therapies. In SCD, a single amino acid substitution in the β-globin chain leads to polymerization of mutant hemoglobin S, impairing erythrocyte rheology and survival. Clinically, erythrocyte abnormalities in SCD manifest in hemolytic anemia and cycles of microvascular vaso-occlusion leading to end-organ ischemia-reperfusion injury and infarction. Vaso-occlusive events and intravascular hemolysis promote inflammation and redox instability that lead to progressive small- and large-vessel vasculopathy. Based on current evidence, the pathobiology of SCD is considered to be a vicious cycle of four major processes, all the subject of active study and novel therapeutic targeting: ( a) hemoglobin S polymerization, ( b) impaired biorheology and increased adhesion-mediated vaso-occlusion, ( c) hemolysis-mediated endothelial dysfunction, and ( d) concerted activation of sterile inflammation (Toll-like receptor 4- and inflammasome-dependent innate immune pathways). These molecular, cellular, and biophysical processes synergize to promote acute and chronic pain and end-organ injury and failure in SCD. This review provides an exhaustive overview of the current understanding of the molecular pathophysiology of SCD, how this pathophysiology contributes to complications of the central nervous and cardiopulmonary systems, and how this knowledge is being harnessed to develop current and potential therapies.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
35
|
Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
36
|
van Beers EJ, van Wijk R. Oxidative stress in sickle cell disease; more than a DAMP squib. Clin Hemorheol Microcirc 2018; 68:239-250. [PMID: 29614635 DOI: 10.3233/ch-189010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sickle cell disease (SCD) is a monogenetic disorder marked by hemolytic anemia and vaso-occlusive complications. The hallmark of SCD is the intracellular polymerization of sickle hemoglobin (HbS) after deoxygenation, and the subsequent characteristic shape change (sickling) of red cells. Vaso-occlusion occurs after endothelial activation, expression of adhesion molecules and subsequent adhesion of leucocytes and sickle erythrocytes to the vascular wall. Here we review how oxidative stress from various sources influences this process. Emerging evidence points towards a dominant mechanism in which innate immune receptors, such as Toll like receptor 4, activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidases to produce reactive oxygen species (ROS) which in turn enables downstream pro-inflammatory signaling and subsequent endothelial activation. By serving as an iron donor for the Fenton reaction, heme radically increases the amount of ROS further, thereby increasing the signal originating from the innate immune receptor and downstream effects of innate immune receptor activation. In SCD this results in the production of pro-inflammatory cytokines, endothelial activation and leucocyte adhesion, and eventually vaso-occlusion. Any intervention to stop this cascade, including Toll like receptor blockade, NADPH oxidase inhibition, ROS reduction, heme scavenging, iron chelation, or anti-adhesion molecule antibodies has been successfully used in pre-clinical studies and holds promise for patients with SCD.
Collapse
Affiliation(s)
- Eduard J van Beers
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Richard van Wijk
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
37
|
Vogel S, Thein SL. Platelets at the crossroads of thrombosis, inflammation and haemolysis. Br J Haematol 2018; 180:761-767. [PMID: 29383704 DOI: 10.1111/bjh.15117] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platelets play a critical role at the interphase of thrombosis and inflammation, key features in haemolysis-associated disorders. Exercising this role requires expression of pattern recognition receptors by platelets, including toll-like receptor 4 (TLR4) and nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3), the latter forming intraplatelet multiprotein inflammasome complexes. Platelets are a potential target of various damage-associated molecular pattern (DAMP) molecules, such as free haem, a degradation by-product of haemoglobin oxidation during haemolysis, and high-mobility group box 1 (HMGB1), a DNA-binding protein released by dying or stressed cells and activated platelets. We have recently identified platelet TLR4, NLRP3, and Bruton tyrosine kinase (BTK) as critical regulators of platelet aggregation and thrombus formation, suggesting that the BTK inhibitor ibrutinib is a potential therapeutic target. Increasing evidence suggests that these and other DAMP-driven signalling mechanisms employed by platelets might be key in mediating inflammation and thrombosis encountered in haemolytic disorders. However, the precise regulatory triggers and their clinical relevance are poorly understood. We provide new insights into these less-well characterised platelet mechanisms, which are potentially targetable in haemolytic disorders.
Collapse
Affiliation(s)
- Sebastian Vogel
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Nwankwo JO, Gremmel T, Gerrits AJ, Mithila FJ, Warburton RR, Hill NS, Lu Y, Richey LJ, Jakubowski JA, Frelinger AL, Chishti AH. Calpain-1 regulates platelet function in a humanized mouse model of sickle cell disease. Thromb Res 2017; 160:58-65. [PMID: 29101791 DOI: 10.1016/j.thromres.2017.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/02/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022]
Abstract
One of the major contributors to sickle cell disease (SCD) pathobiology is the hemolysis of sickle red blood cells (RBCs), which release free hemoglobin and platelet agonists including adenosine 5'-diphosphate (ADP) into the plasma. While platelet activation/aggregation may promote tissue ischemia and pulmonary hypertension in SCD, modulation of sickle platelet dysfunction remains poorly understood. Calpain-1, a ubiquitous calcium-activated cysteine protease expressed in hematopoietic cells, mediates aggregation of platelets in healthy mice. We generated calpain-1 knockout Townes sickle (SSCKO) mice to investigate the role of calpain-1 in steady state and hypoxia/reoxygenation (H/R)-induced sickle platelet activation and aggregation, clot retraction, and pulmonary arterial hypertension. Using multi-electrode aggregometry, which measures platelet adhesion and aggregation in whole blood, we determined that steady state SSCKO mice exhibit significantly impaired PAR4-TRAP-stimulated platelet aggregation as compared to Townes sickle (SS) and humanized control (AA) mice. Interestingly, the H/R injury induced platelet hyperactivity in SS and SSCKO, but not AA mice, and partially rescued the aggregation defect in SSCKO mice. The PAR4-TRAP-stimulated GPIIb-IIIa (αIIbβ3) integrin activation was normal in SSCKO platelets suggesting that an alternate mechanism mediates the impaired platelet aggregation in steady state SSCKO mice. Taken together, we provide the first evidence that calpain-1 regulates platelet hyperactivity in sickle mice, and may offer a viable pharmacological target to reduce platelet hyperactivity in SCD.
Collapse
Affiliation(s)
- Jennifer O Nwankwo
- Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Thomas Gremmel
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Anja J Gerrits
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Farha J Mithila
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Rod R Warburton
- Division of Pulmonary, Critical Care and Sleep Medicine, Tufts Medical Center, Boston, MA, USA
| | - Nicholas S Hill
- Division of Pulmonary, Critical Care and Sleep Medicine, Tufts Medical Center, Boston, MA, USA
| | - Yunzhe Lu
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Lauren J Richey
- Division of Laboratory Animal Medicine, Tufts University, Boston, MA, USA
| | | | - Andrew L Frelinger
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Athar H Chishti
- Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
39
|
Balandya E, Reynolds T, Obaro S, Makani J. Alteration of lymphocyte phenotype and function in sickle cell anemia: Implications for vaccine responses. Am J Hematol 2016; 91:938-46. [PMID: 27237467 DOI: 10.1002/ajh.24438] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 02/07/2023]
Abstract
Individuals with sickle cell anemia (SCA) have increased susceptibility to infections, secondary to impairment of immune function. Besides the described dysfunction in innate immunity, including impaired opsonization and phagocytosis of bacteria, evidence of dysfunction of T and B lymphocytes in SCA has also been reported. This includes reduction in the proportion of circulating CD4+ and CD8+ T cells, reduction of CD4+ helper: CD8+ suppressor T cell ratio, aberrant activation and dysfunction of regulatory T cells (Treg ), skewing of CD4+ T cells towards Th2 response and loss of IgM-secreting CD27 + IgM(high) IgD(low) memory B cells. These changes occur on the background of immune activation characterized by predominance of memory CD4+ T cell phenotypes, increased Th17 signaling and elevated levels of C-reactive protein and pro-inflammatory cytokines IL-6 and TNF-α, which may affect the immunogenicity and protective efficacy of vaccines available to prevent infections in SCA. Thus, in order to optimize the use of vaccines in SCA, a thorough understanding of T and B lymphocyte functions and vaccine reactivity among individuals with SCA is needed. Studies should be encouraged of different SCA populations, including sub-Saharan Africa where the burden of SCA is highest. This article summarizes our current understanding of lymphocyte biology in SCA, and highlights areas that warrant future research. Am. J. Hematol. 91:938-946, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Emmanuel Balandya
- Muhimbili University of Health and Allied Sciences (MUHAS); P.O. Box 65001 Dar-es-Salaam Tanzania
| | - Teri Reynolds
- Muhimbili University of Health and Allied Sciences (MUHAS); P.O. Box 65001 Dar-es-Salaam Tanzania
- University of San Francisco, California (UCSF); 505 Parnassus Ave. San Francisco CA 94143 United States
| | - Stephen Obaro
- University of Nebraska Medical Center (UNMC), 982162 Nebraska Medical Center; Omaha Nebraska 68198-2162 United States
- University of Abuja Teaching Hospital; Gwagwalada, P.M.B 228 Abuja Nigeria
| | - Julie Makani
- Muhimbili University of Health and Allied Sciences (MUHAS); P.O. Box 65001 Dar-es-Salaam Tanzania
| |
Collapse
|
40
|
Pitanga TN, Oliveira RR, Zanette DL, Guarda CC, Santiago RP, Santana SS, Nascimento VML, Lima JB, Carvalho GQ, Maffili VV, Carvalho MOS, Alcântara LCJ, Borges VM, Goncalves MS. Sickle red cells as danger signals on proinflammatory gene expression, leukotriene B4 and interleukin-1 beta production in peripheral blood mononuclear cell. Cytokine 2016; 83:75-84. [PMID: 27045344 DOI: 10.1016/j.cyto.2016.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
This study tested the hypothesis that sickle red blood cell (SS-RBC) induce Toll-like receptors (TLR) and Nod-like receptor family, pyrin domain containing 3 (NLRP3)- inflammasome expression in peripheral blood mononuclear cells (PBMC). TLR and NLRP3 inflammasome could contribute to the maintenance of the inflammatory status in sickle cell anemia (SCA) patients, since SS-RBC act as danger signals activating these pathways. In this study, first, we evaluated TLR (2, 4, 5 and 9), NLRP3, Caspase-1, interleukin (IL)-1β and IL-18 expression in PBMC freshly isolated from SCA patients (SS-PBMC) in comparison with PBMC from healthy individuals (AA-PBMC). In the second moment, we investigated whether SS-RBC could interfere with the expression of these molecules in PBMC from healthy donor, in the absence or presence of hydroxyurea (HU) in vitro. TLRs and NLRP3 inflammasome expression were investigated by qPCR. IL-1β, Leukotriene-B4 (LTB4) and nitrite production were measured in PBMC (from healthy donor) culture supernatants. TLR2, TLR4, TLR5, NLRP3 and IL-1β were highly expressed in SS-PBMC when compared to AA-PBMC. Additionally, SS-RBC induced TLR9, NLRP3, Caspase-1, IL-1β and IL-18 expression and induced IL-1β, LTB4 and nitrite production in PBMC cultures. HU did not prevent TLR and NLRP3 inflammasome expression, but increased TLR2 and IL-18 expression and reduced nitrite production. In conclusion, our data suggest that TLR and inflammasome complexes may be key inducers of inflammation in SCA patients, probably through SS-RBC; also, HU does not prevent NLRP3 inflammasome- and TLR-dependent inflammation, indicating the need to develop new therapeutic strategies to SCA patients that act with different mechanisms of those observed for HU.
Collapse
Affiliation(s)
- Thassila N Pitanga
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Instituto de Ciências da Saúde (ICS), Universidade Federal da Bahia (UFBA), 40110902 Salvador, Bahia, Brazil
| | - Ricardo R Oliveira
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Dalila L Zanette
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Caroline C Guarda
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Rayra P Santiago
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Sanzio S Santana
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Valma M L Nascimento
- Fundação de Hematologia e Hemoterapia da Bahia (HEMOBA), 40286240 Salvador, Bahia, Brazil
| | - Jonilson B Lima
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Graziele Q Carvalho
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Vitor V Maffili
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Magda O S Carvalho
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Hospital Universitário Professor Edgard Santos (HUPES), UFBA, 40110060 Salvador, Bahia, Brazil
| | - Luiz C J Alcântara
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Valéria M Borges
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Marilda S Goncalves
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Faculdade de Farmácia, UFBA, 40170115 Salvador, Bahia, Brazil.
| |
Collapse
|
41
|
Yu B, Li X, Wan Q, Han W, Deng C, Guo C. High-Mobility Group Box-1 Protein Disrupts Alveolar Elastogenesis of Hyperoxia-Injured Newborn Lungs. J Interferon Cytokine Res 2016; 36:159-68. [PMID: 26982166 DOI: 10.1089/jir.2015.0080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although high-mobility group box-1 (HMGB1) levels in tracheal aspirates are associated with the pathological features of bronchopulmonary dysplasia (BPD), the role of HMGB1 in the terminal stage of abnormal alveologenesis has not yet been understood. In this study, we addressed the role of HMGB1 in the elastogenesis disruption in the lungs of newborn mice with BPD. We found that elevations of whole lung HMGB1 level were associated with impaired alveolar development and aberrant elastin production in 85% O2-exposed lungs. HMGB1 neutralizing antibody attenuated the structural disintegration developed in hyperoxia-damaged lungs. Furthermore, HMGB1 inhibition rescued the neutrophil influx in hyperoxia-injured lung and partially abolished the mRNA level of the proinflammatory mediators, interleukin (IL)-1β and transforming growth factor (TGF)-β1. These data suggested that pulmonary HMGB1 plays an important role in the disruption of elastogenesis in the terminal stage of lung development through reduced pulmonary inflammatory response.
Collapse
Affiliation(s)
- Benli Yu
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Xiaoyu Li
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Qiufeng Wan
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Wenli Han
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,3 Department of Pharmacology, Chongqing Medical University , Chongqing, P.R. China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,4 Department of Hepatology and Liver Transplantation Center, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| |
Collapse
|
42
|
Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 2016; 127:801-9. [PMID: 26758915 DOI: 10.1182/blood-2015-09-618538] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023] Open
Abstract
Sickle cell disease (SCD) is a severe genetic blood disorder characterized by hemolytic anemia, episodic vaso-occlusion, and progressive organ damage. Current management of the disease remains symptomatic or preventative. Specific treatment targeting major complications such as vaso-occlusion is still lacking. Recent studies have identified various cellular and molecular factors that contribute to the pathophysiology of SCD. Here, we review the role of these elements and discuss the opportunities for therapeutic intervention.
Collapse
|
43
|
Rankine-Mullings A, Reid ME, Moo Sang M, Richards-Dawson MA, Knight - Madden JM. A Retrospective Analysis of the Significance of Haemoglobin SS and SC in Disease Outcome in Patients With Sickle Cell Disease and Dengue Fever. EBioMedicine 2015; 2:937-41. [PMID: 26425701 PMCID: PMC4563127 DOI: 10.1016/j.ebiom.2015.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 11/30/2022] Open
Abstract
Background Little is known about the significance of haemoglobin genotype in dengue fever severity. This study was undertaken to determine the case fatality ratio and the impact of genotype in patients with sickle cell disease and confirmed dengue fever. Methods This retrospective analysis included 40 patients with confirmed dengue and sickle cell disease, during the study period (2010–2012). Findings There was a significantly higher case fatality ratio, 12.5% among patients with either haemoglobin SC disease or homozygous SS disease when compared to that of the general population 0.41% (p < 0.0001). The unadjusted odds of dying among those with haemoglobin SC disease compared with the group with homozygous SS disease was OR = 4.4 (95% CI 0.6 to 31.7). The predictors of mortality independent of sickle cell disease genotype were haemoglobin concentration at presentation OR = 0.57 (95% CI, 0.35 to 0.94) and the change in haemoglobin concentration from steady state OR = 0.59 (95% CI, 0.37 to 0.94). Adjusting for haemoglobin concentration at presentation increased the risk of death for the SC genotype relative to SS genotype OR = 13.4 (95% CI 1.1 to 160.3). Interpretation The risk of fatal dengue may be higher among patients with a relatively mild genotype (haemoglobin SC). Dengue Fever case fatality ratio was 12.5%( p < 0.0001) in persons with Sickle Cell Disease. The odds of dying from dengue and SC disease was 4.4 (95% CI 0.6 to 31.7).
Collapse
Affiliation(s)
- Angela Rankine-Mullings
- Sickle Cell Unit, Tropical Medicine Research Institute, University of the West Indies, Mona Kingston 7, Jamaica
- Corresponding author.
| | - Marvin E. Reid
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona Kingston 7, Jamaica
| | - Michelle Moo Sang
- Sickle Cell Unit, Tropical Medicine Research Institute, University of the West Indies, Mona Kingston 7, Jamaica
| | | | - Jennifer M. Knight - Madden
- Sickle Cell Unit, Tropical Medicine Research Institute, University of the West Indies, Mona Kingston 7, Jamaica
| |
Collapse
|