1
|
Bordat C, Nader E, Connes P, Joly P, Poutrel S, Perez A, Niesor E. Effect of dalcetrapib, voxelotor and their combination on red blood cell deformability and sickling in sickle cell disease. Clin Hemorheol Microcirc 2025:13860291251320332. [PMID: 40255066 DOI: 10.1177/13860291251320332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The aim of the present study was to test the effects of dalcetrapib and voxelotor on red blood cells (RBC) of sickle cell patients. Oxygen gradient ektacytometry was performed to measure RBC deformability in normoxia and hypoxia, as well as the propensity of RBC to sickle. Voxelotor and dalcetrapib reduced the propensity of RBC to sickle under deoxygenation and increased RBC deformability in hypoxia. Dalcetrapib did not affect the affinity of hemoglobin S (HbS) to oxygen. Combining the two molecules caused greater RBC rheological improvement. Our findings suggest that dalcetrapib could block HbS polymerization without affecting HbS oxygen affinity.
Collapse
Affiliation(s)
- Claire Bordat
- Hartis Pharma SA Nyon, Nyon, Switzerland
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| | - Elie Nader
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| | - Philippe Connes
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
| | - Philippe Joly
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-Site, Hospices Civils de Lyon, Lyon, France
| | - Solene Poutrel
- Laboratory LIBM EA7424, Vascular Biology and Red Blood Cell Team, University of Lyon, Lyon, France
- Service de Médecine Interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Anne Perez
- Hartis Pharma SA Nyon, Nyon, Switzerland
| | | |
Collapse
|
2
|
Shen D, Ju H, Wang H, Wang X, Li G. The clinic application of mNGS and ENA-78 assays to identify intra-amniotic infection/inflammation. Front Cell Infect Microbiol 2025; 15:1510671. [PMID: 40256453 PMCID: PMC12006116 DOI: 10.3389/fcimb.2025.1510671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025] Open
Abstract
Objective The objective of this study is to explore whether metagenomic next-generation sequencing (mNGS) and Epithelial Neutrophil Activating Peptide-78 (ENA-78) assays in the amniotic fluid (AF) of patients with preterm labor (PTL) could be employed for diagnosing intra-amniotic infection/inflammation (IAI/I) and predict the outcomes of emergency cerclage in women with cervical insufficiency(CI). Methods AF samples from 40 patients were subjected to PTL were subjected to mNGS and microbial culture to diagnose intra-amniotic infection known as microbial invasion of the amniotic cavity (MIAC); ELISA was used to analyze ENA-78 levels for prediction of intra-amniotic inflammation (IAI). Pregnancy outcomes were compared, the predictive performance of mNGS and ENA-78 were assessed to evaluate the efficacy of emergency cervical cerclage. Results The diagnosis rate of MIAC was higher with mNGS (17.5%) compared to microbial culture (2.5%). AF ENA-78 levels were significantly higher in IAI patients than in non-IAI/I patients. ENA-78 demonstrated certain accuracy in identifying IAI, with sensitivity and specificity of 73.3% and 100%, respectively. Compared with non-IAI/I patients, patients with MIAC or IAI exhibited poor pregnancy outcomes after cervical cerclage. Conclusions mNGS and ENA-78 assays are valuable means for assessing the state of infection/inflammation in the amniotic cavity and predicting the outcomes of emergency cerclage.
Collapse
Affiliation(s)
- Di Shen
- Department of Obstetrics and Gynaecology, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
- Department of Obstetrics and Gynecology, Liao Cheng People’s Hospital, Liaocheng, China
| | - Hui Ju
- Department of Obstetrics and Gynaecology, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Hongying Wang
- Department of Obstetrics and Gynecology, Liao Cheng People’s Hospital, Liaocheng, China
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guangzhen Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
3
|
Caruccio L, Byrne K, Stroncek DF. Universal new blood cell elution method with extensive phenotyping. Transfus Med 2025; 35:151-167. [PMID: 39923753 PMCID: PMC11999790 DOI: 10.1111/tme.13126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/22/2024] [Accepted: 01/19/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND/OBJECTIVES No erythrocyte elution method developed is uniformly successful or allows elution/phenotyping together. We previously developed an elution method using deionised formamide. We modified it to be universal for various cell types and call it modified formamide-method (Fm-method). It also preserves cells for phenotyping after elution. MATERIALS AND METHODS Fm-method reagent contains deionised formamide, buffered high salt, EDTA, TE. Elution-reagent is removed by column centrifugation. Blood samples were used for development and validation. Results compared to commercial/common antibody elution/phenotyping methods. RESULTS Fm-method eluted antibodies, complement, other proteins, and nucleic acids and works with erythrocytes, leukocytes, other cells. It worked better than commercial kits used for elution/phenotyping with no false positives/negatives. It did not denature Kell and Lewis antigens and could be repeated as needed on samples to recover more antibodies and clean cells for phenotyping. Western blotting, PAGE and FCM demonstrated eluted proteins were not degraded and cells remained intact. CONCLUSION Fm-method is excellent for elution and phenotyping and permits elution and phenotyping with one method and sample. It is useful for studies of various bound molecules and cell surface structures. It should be possible to elute various simple and complex carbohydrates as well. The Fm-method is efficient, inexpensive, scalable, uses common reagents. It should have excellent applications in various clinical, research, commercial settings.
Collapse
Affiliation(s)
- Lorraine Caruccio
- VA San Diego Healthcare System, San Diego, 92161-0002, United States
| | - Karen Byrne
- National Institutes of Health, Clinical Center, Bethesda, 20892, United States
| | - David F. Stroncek
- National Institutes of Health, Clinical Center, Bethesda, 20892, United States
| |
Collapse
|
4
|
Abramson Z, Olanrewaju A, Kang G, Olufadi Y, Chen PL, Rai P, Heitzer AM, Takemoto CM, Bashir A, Akil N, Hankins JS. Hydroxyurea Therapy and Sleep-Disordered Breathing in Children With Sickle Cell Disease. Pediatr Blood Cancer 2025; 72:e31531. [PMID: 39895429 DOI: 10.1002/pbc.31531] [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: 05/14/2024] [Revised: 12/03/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025]
Abstract
Bidirectional relationships exist among sickle cell disease (SCD), sleep-disordered breathing (SDB), and hydroxyurea therapy, and this complex interplay poses challenges to research attempting to elucidate causal mechanisms and determine therapeutic targets. We therefore set out to study all three components of this clinical triad simultaneously (SCD, SDB, and hydroxyurea therapy), documenting correlations and suggesting underlying pathophysiological mechanisms. Hydroxyurea therapy was associated with improved SDB and with decreased inflammation, implicating the anti-inflammation effects of hydroxyurea as a plausible mechanism driving these correlations. These results underscore the need for longitudinal studies to elucidate causal relationships.
Collapse
Affiliation(s)
- Zachary Abramson
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ayobami Olanrewaju
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yunusa Olufadi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Pei-Lin Chen
- Longitudinal Clinical Cohort of Sickle Cell Clinical Research and Intervention Program, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Parul Rai
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Andrew M Heitzer
- Department of Psychology and Behavioral Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Clifford M Takemoto
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ahsan Bashir
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Division of Pediatric Pulmonary and Sleep Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Nour Akil
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Division of Pediatric Pulmonary and Sleep Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Jane S Hankins
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
5
|
Romanello KS, da Silva JPMDO, Torres FF, Teixeira KKL, Domingos IDF, Arcanjo GDS, Martins DAP, Araujo ADS, Bezerra MAC, Malavazi I, da Silva DGH, da Cunha AF. Unraveling the multifaceted roles of peroxiredoxins in sickle cell anemia: implications in redox and inflammation adaptations. Ann Hematol 2025:10.1007/s00277-025-06294-1. [PMID: 40085210 DOI: 10.1007/s00277-025-06294-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
Sickle cell anemia (SCA) presents a complex interplay of factors, with the production of high levels of reactive oxygen species (ROS) and the chronic inflammatory process leading to chronic oxidative stress. In this context, efficient action of antioxidant systems becomes crucial, with particular emphasis on peroxiredoxins (PRDXs) due to their abundance and vital roles. Our primary objective was to establish associations between gene and protein expression of PRDXs 1, 2, and 6, as well as their reducers TRX1, TRXR1, and SRX1, with the characteristic hyperoxidative status observed in SCA patients. Concomitantly, we assessed the production of other essential antioxidant enzymes (SOD1, CAT, and GPX1) in reticulocytes and erythrocytes and explored mRNA levels of the NRF2/KEAP1/PKCδ complex. Our comprehensive analysis revealed a ∼ 3-fold elevation in ROS levels in erythrocytes of patients compared to healthy individuals. However, the NRF2/KEAP1/PKCδ complex exhibited a significant reduction in gene expression, hinting that another transcription factor may regulate the antioxidant response among SCA patients. In addition, the pattern of increased transcript levels of antioxidants in SCA patients was not associated with their protein levels, indicating a possible degradation by proteasome. The protein content of PRDX2 showed a significant reduction, indicating an increased vulnerability of these cells to oxidative damage. Intriguingly, both PRDXs 1 and 2 exhibited significant increases in the plasma of SCA patients, indicating that, besides their well-known intracellular antioxidant role, these enzymes may also play a vital extracellular role in modulating inflammation in these individuals. Our findings unveil novel insights into the redox metabolism adaption of erythroid cells in response to the presence of HbS in homozygosity, thus, into the complex SCA pathophysiology. Moreover, our study reveals the simultaneous presence of both PRDXs 1 and 2 in the plasma of these patients, thereby offering valuable implications for potential prognostic and therapeutic avenues.
Collapse
Affiliation(s)
- Karen Simone Romanello
- Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos (UFSCar), São Carlos, Brazil
| | - João Pedro Maia de Oliveira da Silva
- Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos (UFSCar), São Carlos, Brazil
| | - Flaviene Felix Torres
- Departamento de Biologia, Universidade Estadual Paulista (UNESP), São José do Rio Preto, Brazil
| | - Karina Kirschner Lopes Teixeira
- Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos (UFSCar), São Carlos, Brazil
| | | | | | | | | | | | - Iran Malavazi
- Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos (UFSCar), São Carlos, Brazil
| | - Danilo Grünig Humberto da Silva
- Departamento de Biologia, Universidade Estadual Paulista (UNESP), São José do Rio Preto, Brazil
- Universidade Federal de Mato Grosso do Sul, Três Lagoas, Brazil
| | - Anderson Ferreira da Cunha
- Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos (UFSCar), São Carlos, Brazil.
- Centro de Ciências Biológicas e da Saúde - Departamento de Genética e Evolução - Laboratório de Bioquímica e Genética Aplicada, Universidade Federal de São Carlos, Rodovia Washington Luís, km 235 - SP-310, Bairro Monjolinho, São Carlos, Brasil.
| |
Collapse
|
6
|
de Ligt LA, Gaartman AE, Konté K, Thakoerdin S, Fijnvandraat K, Kuijpers TW, van Bruggen R, Biemond BJ, Nur E. Plasma inflammatory and angiogenic protein profiling of patients with sickle cell disease. Br J Haematol 2025; 206:954-964. [PMID: 39743683 PMCID: PMC11886948 DOI: 10.1111/bjh.19970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
In this study, we aimed to explore the inflammatory and angiogenic pathways in sickle cell disease (SCD). We used proximity extension assay technology (Olink) to measure 92 plasma proteins involved in inflammation and angiogenesis. Plasma samples were collected from 57 SCD patients (sickle cell anaemia/HbS-β0 thalassaemia-thalassaemia) in steady-state and 13 healthy ethnicity-matched healthy controls (HCs). From 15 patients, paired samples were collected during both steady-state and vaso-occlusive episodes (VOEs) and from 23 SCD patients longitudinal samples were collected before and after treatment with either voxelotor (n = 10), hydroxyurea (n = 8) or allogeneic haematopoietic stem-cell transplantation (n = 5). Fifty plasma proteins were differentially expressed in steady-state SCD patients as compared to HC. These included proteins involved in angiogenesis (i.e. ANGPT1, ANGPT2 and VEGFA), the IL-18 signalling pathway (i.e. IL-6, IL-10, IL-18), T-cell activation (i.e. LAG3, PDCD1) and natural killer (NK)-cell activation (CD244, NCR1, GZMB). While proteins involved in angiogenesis and the IL-18 signalling pathway were further upregulated during VOE, levels of several proteins involved in the IL-18 pathway, T-cell and NK-cell activation and angiogenesis, restored towards levels detected in HCs after curative or disease-modifying treatment. These findings might contribute to a better understanding of SCD pathophysiology and identifying potential new targets for therapeutic interventions.
Collapse
Affiliation(s)
- L. A. de Ligt
- Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamthe Netherlands
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Department of Pediatric HematologyEmma Children's Hospital, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - A. E. Gaartman
- Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamthe Netherlands
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - K. Konté
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - S. Thakoerdin
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - K. Fijnvandraat
- Department of Pediatric HematologyEmma Children's Hospital, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - T. W. Kuijpers
- Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamthe Netherlands
- Department of Pediatric ImmunologyEmma Children's Hospital, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - R. van Bruggen
- Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamthe Netherlands
| | - B. J. Biemond
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - E. Nur
- Department of Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdamthe Netherlands
- Department of HematologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
7
|
Žoldáková M, Novotný M, Khakurel KP, Žoldák G. Hemoglobin Variants as Targets for Stabilizing Drugs. Molecules 2025; 30:385. [PMID: 39860253 PMCID: PMC11767434 DOI: 10.3390/molecules30020385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Hemoglobin is an oxygen-transport protein in red blood cells that interacts with multiple ligands, e.g., oxygen, carbon dioxide, carbon monoxide, and nitric oxide. Genetic variations in hemoglobin chains, such as those underlying sickle cell disease and thalassemias, present substantial clinical challenges. Here, we review the progress in research, including the use of allosteric modulators, pharmacological chaperones, and antioxidant treatments, which has begun to improve hemoglobin stability and oxygen affinity. According to UniProt (as of 7 August 2024), 819 variants of the α-hemoglobin subunit and 771 variants of the β-hemoglobin subunit have been documented, with over 116 classified as unstable. These data demonstrate the urgent need to develop variant-specific stabilizing options. Beyond small-molecule drugs/binders, novel protein-based strategies-such as engineered hemoglobin-binding proteins (including falcilysin, llama-derived nanobodies, and α-hemoglobin-stabilizing proteins)-offer promising new options. As our understanding of hemoglobin's structural and functional diversity grows, so does the potential for genotype-driven approaches. Continued research into hemoglobin stabilization and ligand-binding modification may yield more precise, effective treatments and pave the way toward effective strategies for hemoglobinopathies.
Collapse
Affiliation(s)
- Miroslava Žoldáková
- Faculty of Science, Pavol Jozef Šafárik University in Košice, Park Angelinum 19, 040 01 Košice, Slovakia
| | - Michal Novotný
- AURORA R&D s.r.o., Mojmírova 12, 040 01 Košice, Slovakia
| | - Krishna P. Khakurel
- Extreme Light Infrastructure ERIC, Za Radnici 835, 25241 Dolni Brezany, Czech Republic
| | - Gabriel Žoldák
- Faculty of Science, Pavol Jozef Šafárik University in Košice, Park Angelinum 19, 040 01 Košice, Slovakia
| |
Collapse
|
8
|
Su S, Bao W, Liu Y, Shi PA, Manwani D, Murakhovskaya I, Campbell-Lee S, Lobo CA, Mendelson A, An X, Zhong H, Yi W, Yazdanbakhsh K. IFN-I promotes T-cell-independent immunity and RBC autoantibodies via modulation of B-1 cell subsets in murine SCD. Blood 2025; 145:334-347. [PMID: 39656114 PMCID: PMC11775509 DOI: 10.1182/blood.2024025175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/11/2024] [Indexed: 01/18/2025] Open
Abstract
ABSTRACT The pathophysiology of sickle cell disease (SCD) is characterized by hemolytic anemia and vaso-occlusion, although its impact on the adaptive immune responses remains incompletely understood. To comprehensibly profile the humoral immune responses, we immunized SCD mice with T-cell-independent (TI) and T-cell-dependent (TD) antigens (Ags). Our study showed that SCD mice have significantly enhanced type 2 TI (TI-2) immune responses in a manner dependent on the level of type I interferons (IFN-I), while maintaining similar or decreased TD immune responses depending on the route of Ag administration. Consistent with the enhanced TI-2 immune responses in SCD mice, the frequencies of B-1b cells (B-1 cells in humans), a major cell type responding to TI-2 Ags, were significantly increased in both the peritoneal cavity and spleens of SCD mice and in the blood of patients with SCD. In support of expanded B-1 cells, elevated levels of anti-red blood cell (anti-RBC) autoantibodies were detected in both SCD mice and patients. Both the levels of TI-2 immune responses and anti-RBC autoantibodies were significantly reduced after IFN-I receptor (IFNAR) antibody blockades and in IFNAR1-deficient SCD mice. Moreover, the alterations of B-1 cell subsets were reversed in IFNAR1-deficient SCD mice, uncovering a critical role for IFN-I in the enhanced TI-2 immune responses and the increased production of anti-RBC autoantibodies by modulating the innate B-1 cell subsets in SCD. Overall, our study provides experimental evidence that the modulation of B-1 cells and IFN-I can regulate TI immune responses and the levels of anti-RBC autoantibodies in SCD.
Collapse
Affiliation(s)
- Shan Su
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Weili Bao
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Yunfeng Liu
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Patricia A. Shi
- Clinical Research in Sickle Cell Disease, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Deepa Manwani
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, NY
| | - Irina Murakhovskaya
- Department of Hematology and Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY
| | | | - Cheryl A. Lobo
- Laboratory of Blood-Borne Parasites, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Avital Mendelson
- Laboratory of Stem Cell Biology and Engineering, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Xiuli An
- Laboratory of Membrane Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Hui Zhong
- Laboratory of Immune Regulation, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Woelsung Yi
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| |
Collapse
|
9
|
Alexandrova-Watanabe A, Abadjieva E, Gartcheva L, Langari A, Ivanova M, Guenova M, Tiankov T, Strijkova V, Krumova S, Todinova S. The Impact of Targeted Therapies on Red Blood Cell Aggregation in Patients with Chronic Lymphocytic Leukemia Evaluated Using Software Image Flow Analysis. MICROMACHINES 2025; 16:95. [PMID: 39858750 PMCID: PMC11767778 DOI: 10.3390/mi16010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
Chronic lymphocytic leukemia (CLL), the most common type of leukemia, remains incurable with conventional therapy. Despite advances in therapies targeting Bruton's tyrosine kinase and anti-apoptotic protein BCL-2, little is known about their effect on red blood cell (RBC) aggregation in blood flow. In this study, we applied a microfluidic device and a newly developed Software Image Flow Analysis to assess the extent of RBC aggregation in CLL patients and to elucidate the hemorheological effects of the commonly applied therapeutics Obinutuzumab/Venetoclax and Ibrutinib. The results revealed that, in RBC samples from untreated CLL patients, complex 3D clusters of large RBC aggregates are formed, and their number is significantly increased compared to healthy control samples. The application of the Obinutuzumab/Venetoclax combination did not affect this aspect of RBCs' rheological behavior. In contrast, targeted therapy with Ibrutinib preserves the aggregation state of CLL RBCs to levels seen in healthy controls, demonstrating that Ibrutinib mitigates the alterations in the rheological properties of RBCs associated with CLL. Our findings highlight the alterations in RBC aggregation in CLL and the impact of different targeted therapies on RBCs' rheological properties, which is critical for predicting the potential complications and side effects of CLL treatments, particularly concerning blood flow dynamics.
Collapse
Affiliation(s)
- Anika Alexandrova-Watanabe
- Institute of Mechanics, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria; (A.A.-W.); (E.A.); (T.T.)
- Center of Competence at Mechatronics and Clean Technologies—MIRACle, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria;
| | - Emilia Abadjieva
- Institute of Mechanics, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria; (A.A.-W.); (E.A.); (T.T.)
- Center of Competence at Mechatronics and Clean Technologies—MIRACle, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria;
| | - Lidia Gartcheva
- National Specialized Hospital for Active Treating of Hematological Diseases, Zdrave Str. 2, 1756 Sofia, Bulgaria; (L.G.); (M.G.)
| | - Ariana Langari
- Center of Competence at Mechatronics and Clean Technologies—MIRACle, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria;
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (M.I.); (S.K.)
| | - Miroslava Ivanova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (M.I.); (S.K.)
| | - Margarita Guenova
- National Specialized Hospital for Active Treating of Hematological Diseases, Zdrave Str. 2, 1756 Sofia, Bulgaria; (L.G.); (M.G.)
| | - Tihomir Tiankov
- Institute of Mechanics, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria; (A.A.-W.); (E.A.); (T.T.)
- Center of Competence at Mechatronics and Clean Technologies—MIRACle, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria;
| | - Velichka Strijkova
- Institute of Optical Materials and Technologies “Acad. Yordan Malinovski”, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 109, 1113 Sofia, Bulgaria;
| | - Sashka Krumova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (M.I.); (S.K.)
| | - Svetla Todinova
- Center of Competence at Mechatronics and Clean Technologies—MIRACle, “Acad. G. Bontchev” Str. 4, 1113 Sofia, Bulgaria;
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (M.I.); (S.K.)
| |
Collapse
|
10
|
Dorneles J, de Menezes Mayer A, Chies JAB. Sickle Cell Anemia and Inflammation: A Review of Stones and Landmarks Paving the Road in the Last 25 Years. Hematol Rep 2025; 17:2. [PMID: 39846606 PMCID: PMC11755431 DOI: 10.3390/hematolrep17010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
A quarter of a century ago, sickle cell disease (SCD) was mainly viewed as a typical genetic disease inherited as a classical Mendelian trait. Therefore, the main focus concerning SCD was on diagnosis, meaning, genotyping, and identification of homozygous and heterozygous individuals carrying the relevant HbS mutant allele. Nowadays, it is well established that sickle cell disease is indeed the result of homozygosis for the HbS variant, although this single feature is not capable of explaining the highly diverse clinical presentation of SCD. In fact, an important feature of SCD is the chronic inflammation that accompanies the sickling of erythrocytes. In this manuscript, we will revisit the early evidence of inflammation in SCD and review what was uncovered during the last 25 years. Here, we describe Sickle cell anemia as a major participant in the history of science. In fact, SCD was the first genetic disease where the causal mutation was identified and is also the first disease for which treatment through genome editing was approved, making this disease a landmark in the road of molecular biology.
Collapse
Affiliation(s)
| | | | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Post Graduation Program in Genetics and Molecular Biology (PPGBM), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (J.D.); (A.d.M.M.)
| |
Collapse
|
11
|
Drosdowski D, Türck P, Tasca S, Rosa GDL, Guzzo EFM, Bianchi SE, Coitinho AS, Carraro CC, Belló-Klein A, de Castro AL, Bassani VL, Araujo ASDR. Impact of blueberry extract on hematological response in phenylhydrazine-induced hemolytic anemia. Hematol Transfus Cell Ther 2025; 47:103744. [PMID: 40014902 PMCID: PMC11910687 DOI: 10.1016/j.htct.2025.103744] [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: 07/12/2024] [Accepted: 11/04/2024] [Indexed: 03/01/2025] Open
Abstract
The objective of this study was to explore the therapeutic effect of blueberries on hematological parameters, oxidative stress, and interleukin-10 levels in acute hemolytic anemia induced by the administration of an intraperitoneal injection of 40 mg/kg phenylhydrazine. Male Wistar rats were divided into three groups: Control, anemia (PHZ), and anemia plus blueberries (PHZ+BB). Blueberries were administered via oral gavage (250 mg/day). The erythrocyte osmotic fragility, splenomegaly, iron metabolism, hematological analysis, reactive oxygens species, sulfhydryl group, and interleukin-10 levels were evaluated. The erythrocyte osmotic fragility (in 0.85% and 0.55% sodium chloride solution) and spleen weight-to-body weight ratio (∼400%) were elevated in the PHZ and PHZ+BB Groups compared to the controls (p-value < 0.05). Increased transferrin and reactive oxygens species levels were found in the PHZ (15%) compared to the Control Group (p-value < 0.05). There was an immune inflammatory response in the PHZ Group due to increases in the total leukocyte (300%), lymphocyte (100%), and neutrophil (400%) counts compared to the Control Group (p-value < 0.05); the PHZ Group showed increased interleukin-10 levels (100%) compared to the Control Group (p-value < 0.05). Blueberries showed a partial protective effect on these parameters, since there were lower neutrophil and lymphocyte counts and diminished interleukin-10 levels in the PHZ+BB Group compared to the PHZ Group (p-value < 0.05). In addition, blueberries increased sulfhydryl group levels (p-value < 0.05). These data suggest a protective role of blueberries against inflammatory response and oxidative stress in an acute hemolytic anemia model.
Collapse
Affiliation(s)
- Daniela Drosdowski
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Patrick Türck
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Silvio Tasca
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | | | - Sara Elis Bianchi
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
12
|
Costa A, Mendes IF, Lage J, Moniz M, Amorim C, Nunes P, Almeida H, Ventura A, Ferreira T, Escobar C. Impact of Red Cell Exchange Transfusion on Inflammatory Markers in Sickle Cell Disease. Mediterr J Hematol Infect Dis 2025; 17:e2025011. [PMID: 39830790 PMCID: PMC11740914 DOI: 10.4084/mjhid.2025.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
Background Red Blood Cell Exchange (RBCX) is a common treatment for pediatric sickle cell disease (SCD). Since inflammation with elevated proinflammatory cytokines plays a crucial role in SCD, this study hypothesized that RBCX might lower these cytokines and aimed to assess the impact of this technique on these markers. Methods Prospective and observational study of pediatric SCD patients (HbSS genotype) enrolled in a chronic RBCX program at a Portuguese Hospital from October 2022 to August 2024. Blood samples were collected before and after RBCX to assess hematological and inflammatory markers. Data were analyzed using SPSSv25 ® (Significance level p < 0.05); Informed consents were obtained. Results Thirty-one children (median age 10 years) were studied: 14 were treatment-naïve, and 17 were previously in a chronic RBCX program. The primary indication for starting the program was cerebrovascular disease prevention (81%). Analysis of 286 RBCXs showed no major adverse events or disease-related hospitalizations. Hemoglobin levels increased by 1.5g/dL post-RBCX; HbS, leukocytes, IL-1, and CRP decreased by 69%, 20%, 21%, and 13%, respectively. Other markers showed no significant changes. IL-1, ferritin, and procalcitonin showed high levels before RBCX; IL-6 showed high levels post-RBCX. Considering only naïve patients, they had higher pre-RBCX IL-1 levels than those with prior RBCX (difference of 22.6 pg/mL); IL-6 increased by 17.3% and IL-1 decreased by 23.9% post-RBCX (p < 0.001). Conclusions RBCX safely reduces HbS, leukocytes, and IL-1 levels, suggesting a modulatory effect on inflammation in SCD patients. Further research is needed to explore cytokine mechanisms in SCD.
Collapse
Affiliation(s)
- Adriana Costa
- Pediatrics Service, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Inês Filipa Mendes
- Pediatrics Service, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Joana Lage
- Pediatrics Service, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Marta Moniz
- Pediatric Intensive Care Unit, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Catarina Amorim
- Pediatric Intensive Care Unit, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Pedro Nunes
- Pediatric Intensive Care Unit, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Helena Almeida
- Pediatric Intensive Care Unit, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Ana Ventura
- Pediatrics Service, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Teresa Ferreira
- Pediatrics Service, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| | - Carlos Escobar
- Pediatric Intensive Care Unit, Hospital Professor Doutor Fernando Fonseca, Unidade Local de Saúde Amadora/Sintra, Portugal
| |
Collapse
|
13
|
Özer Ö, Doğan L, Baysal Z, Basir H, Çıftçı AT, Eröz P, Güçlü ES. Evaluation of peripheral blood inflammatory biomarkers in sickle cell disease with and without retinopathy. Graefes Arch Clin Exp Ophthalmol 2024; 262:3787-3796. [PMID: 38976013 PMCID: PMC11608169 DOI: 10.1007/s00417-024-06569-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND The aim of this study was to evaluate the clinical significance of blood-cell associated inflammation markers in patients with sickle cell disease (SCD) and sickle cell retinopathy (SCR). METHODS Neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), monocyte to lymphocyte ratio (MLR), systemic immune inflammation index (SIII), systemic inflammation response index (SIRI), systemic inflammation modulation index (SIMI) and aggregate systemic inflammation index (AISI) were calculated. This study included 45 healthy controls (Group 1) and 100 SCD (Group 2). Patients in Group 2 were then divided into two groups: without SCR (Group 3) and with SCR (Group 4), and patients with SCR (Group 4) were further divided into two groups: non-proliferative sickle cell retinopathy (NPSCR) (Group 5) and proliferative sickle cell retinopathy (PSCR) (Group 6). RESULTS The mean values for NLR, PLR, SIII, SIRI, AISI, and SIMI were significantly higher in Group 2 compared to Group 1 (p = 0.011 for NLR, p = 0.004 for SIII, and p < 0.001 for others). Furthermore, AISI and SIMI parameters demonstrated statistically significant discriminatory power to distinguish Group 5 from Group 6 (p = 0.0016 and p = 0.0006, respectively). CONCLUSION Given the critical role of inflammatory mechanisms in the pathogenesis of SCD and its related complications, the assessment of blood-cell-associated inflammatory markers may present a pragmatic and advantageous approach to the clinical oversight and therapeutic intervention of SCD.
Collapse
Affiliation(s)
- Ömer Özer
- Department of Ophthalmology, Niğde Ömer Halisdemir University, Niğde, 51240, Turkey.
| | - Levent Doğan
- Department of Ophthalmology, Niğde Ömer Halisdemir University, Niğde, 51240, Turkey
| | - Zeki Baysal
- Department of Ophthalmology, Niğde Ömer Halisdemir University, Niğde, 51240, Turkey
| | - Hakan Basir
- Clinic of Internal Medicine, Gülnar State Hospital, Mersin, Turkey
| | - Ali Türker Çıftçı
- Department of Biostatistics and Medical Informatics, Niğde Ömer Halisdemir University, Niğde, Turkey
| | - Pınar Eröz
- Clinic of Ophthalmology, Tarsus State Hospital, Mersin, Turkey
| | | |
Collapse
|
14
|
DeVeaux SA, Vyshnya S, Propsom K, Gbotosho OT, Singh AS, Horning RZ, Sharma M, Jegga AG, Niu L, Botchwey EA, Hyacinth HI. Neuroinflammation underlies the development of social stress induced cognitive deficit in male sickle cell mice. Exp Biol Med (Maywood) 2024; 249:10361. [PMID: 39629138 PMCID: PMC11612828 DOI: 10.3389/ebm.2024.10361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
Cognitive deficit is a debilitating complication of sickle cell disease (SCD), with a multifactorial etiopathogenesis. Here we show that neuroinflammation and dysregulation in lipidomics and transcriptomics profiles are major underlying mechanisms of social stress-induced cognitive deficit in SCD. Male Townes sickle cell (SS) mice and controls (AA) were exposed to social stress using the repeat social defeat (RSD) paradigm concurrently with or without treatment with minocycline. Mice were tested for cognitive deficit using novel object recognition and fear conditioning tests. SS mice exposed to RSD without treatment had worse performance on cognitive tests compared to SS mice exposed to RSD with treatment or to AA controls, irrespective of their RSD or treatment disposition. Additionally, compared to SS mice exposed to RSD with treatment, SS mice exposed to RSD without treatment had significantly more cellular evidence of neuroinflammation coupled with a significant shift in the differentiation of neural progenitor cells towards astrogliogenesis. Additionally, brain tissue from SS mice exposed to RSD was significantly enriched for genes associated with blood-brain barrier dysfunction, neuron excitotoxicity, inflammation, and significant dysregulation in sphingolipids important to neuronal cell processes. We demonstrate in this study that social stress induces cognitive deficit in SS mice, concurrently with neuroinflammation and lipid dysregulation.
Collapse
Affiliation(s)
- S’Dravious A. DeVeaux
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, United States
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, United States
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Katherine Propsom
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Asem S. Singh
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Robert Z. Horning
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Mihika Sharma
- Division of Biomedical Informatics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH, United States
| | - Anil G. Jegga
- Division of Biomedical Informatics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH, United States
| | - Liang Niu
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, United States
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
15
|
Majrashi NA, Hendi AM, Dhayihi TM, Khamesi AM, Masmali MA, Hakami KJ, Alyami AS, Alwadani B, Ageeli WA, Madkhali Y, Hakamy A, Refaee TA. Associations of haematological and inflammatory biomarkers with brain volume in patients with sickle cell anaemia: A cross-sectional retrospective study. Trop Med Int Health 2024. [PMID: 39510829 DOI: 10.1111/tmi.14056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Sickle cell disease is a genetic disorder characterised by abnormal haemoglobin production. This study aims to investigate the associations between haematological and inflammatory biomarkers and brain volumes in patients with sickle cell anaemia and compare brain structure between patients with sickle cell anaemia and healthy controls. This retrospective cross-sectional study included 130 participants (70 sickle cell anaemia patients and 60 healthy controls) who underwent brain MRI examinations at King Fahad Central Hospital between January 2010 and October 2022. Demographic data and haematological and inflammatory biomarkers were collected to examine their relationships with brain volumes. Brain volumes were measured using FreeSurfer. Specific haematological and inflammatory biomarkers were correlated with brain volume in patients with sickle cell anaemia, p < 0.05. Sickle cell anaemia patients exhibited smaller volumes in the brainstem, corpus callosum and amygdala compared to healthy controls. Males had significantly higher iron levels (p < 0.001) and larger various brain structure volumes (p < 0.05) than females. This study demonstrates significant associations between specific biomarkers and brain volume in sickle cell anaemia patients, underscoring the importance of monitoring these biomarkers for early detection and management of neurological complications in sickle cell anaemia.
Collapse
Affiliation(s)
- Naif A Majrashi
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ali M Hendi
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Department of Radiology, Faculty of Medicine, Jazan university, Jazan, Saudi Arabia
| | - Turki M Dhayihi
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Department of Radiology, Faculty of Medicine, Jazan university, Jazan, Saudi Arabia
| | - Abdullah M Khamesi
- Radiology Department, Jazan Specialist Hospital, Jazan Health Cluster, Jazan, Saudi Arabia
| | - Mohammed A Masmali
- Radiology Department, King Fahad Central Hospital, Ministry of Health, Jazan Health Affairs, Jazan, Saudi Arabia
| | - Khalid J Hakami
- Radiology Department, King Fahad Central Hospital, Ministry of Health, Jazan Health Affairs, Jazan, Saudi Arabia
| | - Ali S Alyami
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Bandar Alwadani
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Wael A Ageeli
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Yahia Madkhali
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ali Hakamy
- Respiratory Therapy Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Turkey A Refaee
- Diagnostic Radiography Technology (DRT) Department, Faculty of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
16
|
Ita MI, Olesen P, Rosing M, Mørk M, Einarsson HB, Riis JJ. Spontaneous Extradural Hematoma in a Sickle Cell Anemia Patient with Hyperinflammation and Thrombotic Microangiopathy Successfully Treated with Eculizumab: A Case Report and Review of the Literature. J Neurol Surg A Cent Eur Neurosurg 2024; 85:625-632. [PMID: 38378045 DOI: 10.1055/a-2271-8772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
BACKGROUND The event of extradural hematoma in the absence of head trauma is a rare central nervous system complication of sickle cell disease. We report here a case of spontaneous extradural hematoma in a patient being treated for sickle cell vasoocclusive crisis complicated by hyperinflammation and thrombotic microangiopathy. The significance of inflammation as an integral component of the pathomechanism of vasoocclusive crisis in patients with sickle cell disease and the role of heme in activating the complement system's alternative pathway are highlighted in this case report. CASE PRESENTATION A teenage patient with sickle cell disease developed a spontaneous right parietal extradural hematoma while receiving treatment for sickle cell vasoocclusive crisis. The concurrent events of hyperinflammation, disseminated intravascular coagulation, hyperhemolysis syndrome, thrombotic microangiopathy, and refractory postoperative bleeding complicated this patient's clinical course after surgical evacuation of extradural hematoma. This patient was subsequently treated with eculizumab and improved in the days following. CONCLUSION Treatment with the anti-C5 monoclonal antibody eculizumab, which targets and inhibits terminal complement system activation, reversed the deleterious cascade of events in this patient with sickle cell disease.
Collapse
Affiliation(s)
- Michael Itak Ita
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Pia Olesen
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Maria Rosing
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Morten Mørk
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | | | - Jens Jakob Riis
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
17
|
Tang MW, Nur E, Van Tuijn CFJ, Biemond BJ. Prevalence of autoimmune diseases in patients with sickle cell disease: a single center retrospective analysis. Haematologica 2024; 109:3735-3738. [PMID: 38546667 PMCID: PMC11532696 DOI: 10.3324/haematol.2023.284552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/21/2024] [Indexed: 11/05/2024] Open
Affiliation(s)
- Man Wai Tang
- Department of Clinical Hematology, Amsterdam University Medical Center, location Academic Medical Center, Amsterdam.
| | - Erfan Nur
- Department of Clinical Hematology, Amsterdam University Medical Center, location Academic Medical Center, Amsterdam, The Netherlands; Department of Blood Cell Research, Sanquin Research, Amsterdam
| | - Charlotte F J Van Tuijn
- Department of Clinical Hematology, Amsterdam University Medical Center, location Academic Medical Center, Amsterdam
| | - Bart J Biemond
- Department of Clinical Hematology, Amsterdam University Medical Center, location Academic Medical Center, Amsterdam
| |
Collapse
|
18
|
Savina Y, Pichon AP, Lemaire L, Howe CA, Ulliel-Roche M, Skinner S, Nader E, Guillot N, Stauffer É, Roustit M, Hancco I, Robach P, Esteve F, Pialoux V, Perger E, Parati G, Ainslie PN, Doutreleau S, Connes P, Verges S, Brugniaux JV. Micro- and macrovascular function in the highest city in the world: a cross sectional study. LANCET REGIONAL HEALTH. AMERICAS 2024; 38:100887. [PMID: 39381083 PMCID: PMC11459627 DOI: 10.1016/j.lana.2024.100887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/04/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024]
Abstract
Background Since vascular responses to hypoxia in both healthy high-altitude natives and chronic mountain sickness (a maladaptive high-altitude pathology characterised by excessive erythrocytosis and the presence of symptoms-CMS) remain unclear, the role of inflammation and oxidative/nitrosative stress on the endothelium-dependent and -independent responses in both the micro- and macrocirculation, in healthy Andeans at different altitudes and in CMS patients, was examined. Methods 94 men were included: 18 lowlanders (LL), 38 healthy highlanders permanently living at 3800 m (n = 21-HL-3800) or in La Rinconada, the highest city in the world (5100-5300 m) (n = 17-HL-5100/No CMS). Moreover, 14 participants with mild (Mild CMS) and 24 with moderate to severe CMS (Mod/Sev CMS) were recruited. All undertook two reactivity tests: i) local thermal hyperaemia (microcirculation) and ii) flow-mediated dilation (macrocirculation). Endothelium-independent function (glyceryl trinitrate) was also assessed only in La Rinconada. Findings Conductance and skin blood flow velocity during the microcirculation test, as well as macrocirculation progressively decreased with altitude (LL > HL-3800 > HL-5100/No CMS). CMS also induced a decrease in macrocirculation (HL-5100/No CMS > Mild CMS = Mod/Sev CMS), while glyceryl trinitrate restored vascular function. Both oxidative stress and nitric oxide metabolites increased with altitude only. Principal component analysis revealed that increasing inflammation with altitude was associated with a progressive decline in both micro- and macrovascular function in healthy highlanders. Interpretation Both micro and macrovascular function are affected by chronic exposure to hypoxia, the latter being further compounded by CMS. Funding The "Fonds de dotation AGIR pour les maladies chroniques", the "Air Liquide Foundation", and the "French National Research Agency".
Collapse
Affiliation(s)
- Yann Savina
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Aurélien P. Pichon
- Laboratory Mobility, Aging & Exercise (MOVE, EA6314), Faculty of Sport Sciences, University of Poitiers, Poitiers, France
| | - Lucas Lemaire
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Connor A. Howe
- Centre for Heart, Lung, and Vascular Health, University of British Columbia, Kelowna, British Columbia, Canada
| | - Mathilde Ulliel-Roche
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
- Grenoble Alpes University Hospital, Grenoble, France
| | - Sarah Skinner
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
- Laboratory of Excellence on Red Blood Cell (GR-Ex), Paris, France
| | - Elie Nader
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
- Laboratory of Excellence on Red Blood Cell (GR-Ex), Paris, France
| | - Nicolas Guillot
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
- Laboratory of Excellence on Red Blood Cell (GR-Ex), Paris, France
| | - Émeric Stauffer
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
- Laboratory of Excellence on Red Blood Cell (GR-Ex), Paris, France
| | - Matthieu Roustit
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
- Grenoble Alpes University Hospital, Grenoble, France
| | - Ivan Hancco
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Paul Robach
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
- National School for Mountain Sports, Site of the National School for Skiing and Mountaineering (ENSA), Chamonix, France
| | - François Esteve
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Vincent Pialoux
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
| | - Elisa Perger
- Istituto Auxologico Italiano, IRCCS, Sleep Disorders Center & Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Milan, Italy
- University of Milano-Bicocca, Milan, Italy
| | - Gianfranco Parati
- Istituto Auxologico Italiano, IRCCS, Sleep Disorders Center & Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Milan, Italy
- University of Milano-Bicocca, Milan, Italy
| | - Philip N. Ainslie
- Centre for Heart, Lung, and Vascular Health, University of British Columbia, Kelowna, British Columbia, Canada
| | - Stéphane Doutreleau
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
- Grenoble Alpes University Hospital, Grenoble, France
| | - Philippe Connes
- Interuniversity Laboratory of Human Movement Biology (LIBM, EA7424), “Red Blood cell and Vascular Biology” team, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
- Laboratory of Excellence on Red Blood Cell (GR-Ex), Paris, France
| | - Samuel Verges
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
- Grenoble Alpes University Hospital, Grenoble, France
| | - Julien V. Brugniaux
- HP2 laboratory, Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
19
|
Sun W, Li J, Zhong J, Feng J, Ye Z, Lin Y, Su W, Zhu S, Li Y, Jia W. Exploring the effect of hydroxyapatite nanoparticle shape on red blood cells and blood coagulation. Nanomedicine (Lond) 2024; 19:2301-2314. [PMID: 39310995 PMCID: PMC11487950 DOI: 10.1080/17435889.2024.2396152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/20/2024] [Indexed: 10/20/2024] Open
Abstract
Aim: In this study, we evaluated the effects of two types of hydroxyapatite (HAP) nanoparticles, sharing the same surface chemistry but differing in shape, on the biological characteristics of plasma, platelets and red blood cells.Materials & methods: Initially, two different shapes (rod-shaped and sphere-shaped) of HAPs were characterized. These HAPs were then co-cultured with plasma and red blood cells to examine their impact on coagulation and hemolysis. The impact of HAPs on white blood cells count in mice were evaluated following gavage and tail vein injection.Results: Sphere-shaped HAP is more likely to adsorb onto platelet surfaces, while rod-shaped HAP is more likely to cause hemolysis. Although there are differences in the in vitro experimental results between sphere-shaped HAP and rod-shaped HAP, both types demonstrate good blood compatibility at a 20 mM concentration. Furthermore, in vivo experiments showed that sphere-shaped nano-HAP induced a more pronounced increase in white blood cell count, suggesting that it may exhibit greater toxicity.Conclusion: While differences exist in the blood compatibility test results between the two HAPs, these differences are minimal, with both results falling within a safe range. Overall, HAP demonstrates excellent blood compatibility.
Collapse
Affiliation(s)
- Weitang Sun
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Jiali Li
- Center Laboratory, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jingbin Zhong
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Jieling Feng
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Zijie Ye
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Yueling Lin
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Wenqi Su
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Shibo Zhu
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| | - Yinghua Li
- Center Laboratory, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Jia
- Institute of Pediatrics, Guangzhou Women & Children’s Medical Center, Guangzhou Medical University, China
| |
Collapse
|
20
|
Grazioli A, Rabin J, Rector RP, Wu ZJ, Burke AP, Sharifai N, Shah A, Taylor BS, Gladwin MT. Venoarterial Extracorporeal Membrane Oxygenation Therapy in Patients with Sickle Cell Disease: Case Series and Review for Intensive Care Physicians. J Intensive Care Med 2024:8850666241260605. [PMID: 39140386 DOI: 10.1177/08850666241260605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Sickle cell disease (SCD) is associated with substantial morbidity and early mortality in afflicted adults. Cardiopulmonary complications that occur at increased frequency in SCD such as pulmonary embolism, pulmonary arterial hypertension, and acute chest syndrome can acutely worsen right ventricular function and lead to cardiogenic shock. Mechanical circulatory support including venoarterial extracorporeal membrane oxygenation (VA ECMO) is being increasingly utilized to treat hemodynamic collapse in various patient populations. However, a paucity of literature exists to guide the use of mechanical circulatory support in adults with SCD where disease-related sequela and unique hematologic aspects of this disorder may complicate extracorporeal therapy and must be understood. Here, we review the literature and describe three cases of adult patients with SCD who developed cardiogenic shock from acute decompensated right heart failure and were treated clinically with VA ECMO. Using an in vitro ECMO system, we investigate a potential increased risk of systemic fat emboli in patients with SCD who may be experiencing vaso-occlusive events with bone marrow involvement given the high-volume shunting of blood from venous to arterial systems with VA ECMO. The purpose of this study is to describe available extracorporeal life support experiences, review potential complications, and discuss the special considerations needed to further our understanding of the utility of VA ECMO in those with SCD.
Collapse
Affiliation(s)
- Alison Grazioli
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Program in Trauma, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph Rabin
- Department of Surgery, Program in Trauma, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Zhongjun J Wu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Allen P Burke
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nima Sharifai
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aakash Shah
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bradley S Taylor
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark T Gladwin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Kaltsas A, Zikopoulos A, Dimitriadis F, Sheshi D, Politis M, Moustakli E, Symeonidis EN, Chrisofos M, Sofikitis N, Zachariou A. Oxidative Stress and Erectile Dysfunction: Pathophysiology, Impacts, and Potential Treatments. Curr Issues Mol Biol 2024; 46:8807-8834. [PMID: 39194738 DOI: 10.3390/cimb46080521] [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: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition affecting men's sexual health, with oxidative stress (OS) having recently been identified as a significant contributing causative factor. This narrative review aims to elucidate the role of OS in the pathophysiology of ED, focusing on impact, mechanisms, and potential therapeutic interventions. Key findings indicate that OS disrupts endothelial function and nitric oxide (NO) signaling, crucial for erectile function. Various sources of reactive oxygen species (ROS) and their detrimental effects on penile tissue are discussed, including aging, diabetes mellitus, hypertension, hyperlipidemia, smoking, obesity, alcohol consumption, psychological stress, hyperhomocysteinemia, chronic kidney disease, and sickle cell disease. Major sources of ROS, such as NADPH oxidase, xanthine oxidase, uncoupled endothelial NO synthase (eNOS), and mitochondrial electron transport, are identified. NO is scavenged by these ROS, leading to endothelial dysfunction characterized by reduced NO availability, impaired vasodilation, increased vascular tone, and inflammation. This ultimately results in ED due to decreased blood flow to penile tissue and the inability to achieve or maintain an erection. Furthermore, ROS impact the transmission of nitrergic neurotransmitters by causing the death of nitrergic neurons and reducing the signaling of neuronal NO synthase (nNOS), exacerbating ED. Therapeutic approaches targeting OS, including antioxidants and lifestyle modifications, show promise in ameliorating ED symptoms. The review underscores the need for further research to develop effective treatments, emphasizing the interplay between OS and vascular health in ED. Integrating pharmacological and non-pharmacological strategies could enhance clinical outcomes for ED patients, advocating for OS management in ED treatment protocols to improve patient quality of life.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | | | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Danja Sheshi
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Magdalena Politis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Efthalia Moustakli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelos N Symeonidis
- Department of Urology II, European Interbalkan Medical Center, 55535 Thessaloniki, Greece
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
22
|
Pavitra E, Acharya RK, Gupta VK, Verma HK, Kang H, Lee JH, Sahu T, Bhaskar L, Raju GSR, Huh YS. Impacts of oxidative stress and anti-oxidants on the development, pathogenesis, and therapy of sickle cell disease: A comprehensive review. Biomed Pharmacother 2024; 176:116849. [PMID: 38823275 DOI: 10.1016/j.biopha.2024.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024] Open
Abstract
Sickle cell disease (SCD) is the most severe monogenic hemoglobinopathy caused by a single genetic mutation that leads to repeated polymerization and depolymerization of hemoglobin resulting in intravascular hemolysis, cell adhesion, vascular occlusion, and ischemia-reperfusion injury. Hemolysis causes oxidative damage indirectly by generating reactive oxygen species through various pathophysiological mechanisms, which include hemoglobin autoxidation, endothelial nitric oxide synthase uncoupling, reduced nitric oxide bioavailability, and elevated levels of asymmetric dimethylarginine. Red blood cells have a built-in anti-oxidant system that includes enzymes like sodium dismutase, catalase, and glutathione peroxidase, along with free radical scavenging molecules, such as vitamin C, vitamin E, and glutathione, which help them to fight oxidative damage. However, these anti-oxidants may not be sufficient to prevent the effects of oxidative stress in SCD patients. Therefore, in line with a recent FDA request that the focus to be placed on the development of innovative therapies for SCD that address the root cause of the disease, there is a need for therapies that target oxidative stress and restore redox balance in SCD patients. This review summarizes the current state of knowledge regarding the role of oxidative stress in SCD and the potential benefits of anti-oxidant therapies. It also discusses the challenges and limitations of these therapies and suggests future directions for research and development.
Collapse
Affiliation(s)
- Eluri Pavitra
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea; 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Rakesh Kumar Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh 495009, India
| | - Vivek Kumar Gupta
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of lungs health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, Munich 85764, Germany
| | - Haneul Kang
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Tarun Sahu
- Department of Physiology, All Indian Institute of Medical Science, Raipur, Chhattisgarh, India
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh 495009, India.
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
23
|
de Ligt LA, Gaartman AE, Biemond BJ, Fijnvandraat K, van Bruggen R, Nur E. Neutrophils in sickle cell disease: Exploring their potential role as a therapeutic target. Am J Hematol 2024; 99:1119-1128. [PMID: 38293835 DOI: 10.1002/ajh.27224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/15/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024]
Abstract
Factors influencing the activation of neutrophils in SCD and the potential neutrophil-mediated ameliorating effects of therapies in SCD.
Collapse
Affiliation(s)
- Lydian A de Ligt
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Aafke E Gaartman
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Bart J Biemond
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Erfan Nur
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Tang MS, Shan H. Current state of gene therapy in sickle cell disease. Vox Sang 2024; 119:521-528. [PMID: 38487952 DOI: 10.1111/vox.13612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 06/12/2024]
Abstract
Sickle cell disease (SCD) is a type of hemoglobinopathy due to an autosomal recessive genetic defect, causing significant red cell sickling, multi-organ damage and long-term severe morbidities. Due to its complicated care and the impact on quality of life, a curative treatment for SCD is highly desirable. In recent years, gene therapy is emerging as a curative option for SCD, where autologous haematopoietic stem cells are collected from SCD patients and genetically modified ex vivo to reduce its sickling tendency before reinfusion. Although still largely investigational, a limited number of gene therapy options have been recently granted approval for SCD patients. Published data are still currently limited, but early studies have so far demonstrated the intended outcomes of less vaso-occlusive crisis and haemolysis. Nonetheless, despite its curative potential, larger clinical trials and longer follow-up period are still necessary to evaluate the safety of this treatment option, especially the risk of unintended genetic modifications. Furthermore, SCD patients frequently have limited access to specialty care; hence, the issues of affordability and accessibility to SCD gene therapy must also be addressed for it to benefit the appropriate patient population.
Collapse
Affiliation(s)
- Mei San Tang
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Pathology, Washington University, St Louis, Missouri, USA
| | - Hua Shan
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
25
|
Tao J, Liu L, Huang X, Tu C, Zhang L, Yang S, Bai Y, Li L, Qin Z. FerrylHb induces inflammation and cell death in grass carp (Ctenopharyngodon idella) hepatocytes. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109474. [PMID: 38513914 DOI: 10.1016/j.fsi.2024.109474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/23/2024]
Abstract
Grass carp hemorrhagic disease is a significant problem in grass carp aquaculture. It releases highly oxidizing hemoglobin (Hb) into tissues, induces rapid autooxidation, and subsequently discharges cytotoxic reactive oxygen species (ROS). However, the mechanism underlying Hb damage to the teleost remains unclear. Here, we employed ferrylHb and heme to incubate L8824 (grass carp liver) cells and quantitatively analyzed the corresponding molecular regulation using the RNA-seq method. Based on the RNA-seq analysis data, after 12 h of incubation of the L8824 cells with ferrylHb, a total of 3738 differentially expressed genes (DEGs) were identified, 1824 of which were upregulated, and 1914 were downregulated. A total of 4434 DEGs were obtained in the heme treated group, with 2227 DEGs upregulated and 2207 DEGs downregulated. KEGG enrichment analysis data revealed that the incubation of ferrylHb and heme significantly activated the pathways related to Oxidative Phosphorylation, Autophagy, Mitophagy and Protein Processing in Endoplasmic Reticulum. The genes associated with NF-κB, autophagy and apoptosis pathways were selected for further validation by quantitative real-time RT-PCR (qRT-PCR). The results were consistent with the RNA-seq data. Taken together, the incubation of Hb and heme induced the molecular regulation of L8824, which consequently led to programmed cell death through multiple pathways.
Collapse
Affiliation(s)
- Junjie Tao
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lihan Liu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Xiaoman Huang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Chenming Tu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Linpeng Zhang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Shiyi Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Yanhan Bai
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lin Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
26
|
Gupta P, Kumar R. Nitric oxide: A potential etiological agent for vaso-occlusive crises in sickle cell disease. Nitric Oxide 2024; 144:40-46. [PMID: 38316197 DOI: 10.1016/j.niox.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Nitric oxide (NO), a vasodilator contributes to the vaso-occlusive crisis associated with the sickle cell disease (SCD). Vascular nitric oxide helps in vasodilation, controlled platelet aggregation, and preventing adhesion of sickled red blood cells to the endothelium. It decreases the expression of pro-inflammatory genes responsible for atherogenesis associated with SCD. Haemolysis and activated endothelium in SCD patients reduce the bioavailability of NO which promotes the severity of sickle cell disease mainly causes vaso-occlusive crises. Additionally, NO depletion can also contribute to the formation of thrombus, which can cause serious complications such as stroke, pulmonary embolism etc. Understanding the multifaceted role of NO provides valuable insights into its therapeutic potential for managing SCD and preventing associated complications. Various clinical trials and studies suggested the importance of artificially induced nitric oxide and its supplements in the reduction of severity. Further research on the mechanisms of NO depletion in SCD is needed to develop more effective treatment strategies and improve the management of this debilitating disease.
Collapse
Affiliation(s)
- Parul Gupta
- ICMR-National Institute of Research in Tribal Health, India
| | - Ravindra Kumar
- ICMR-National Institute of Research in Tribal Health, India.
| |
Collapse
|
27
|
Ansong-Ansongton YON, Adamson TD. Computing Sickle Erythrocyte Health Index on quantitative phase imaging and machine learning. Exp Hematol 2024; 131:104166. [PMID: 38246310 DOI: 10.1016/j.exphem.2024.104166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Sickle cell disease (SCD) is a genetic disorder characterized by abnormal hemoglobin and deformation of red blood cells (RBCs), leading to complications and reduced life expectancy. This study developed an in vitro assessment, the Sickle Erythrocyte Health Index, using quantitative phase imaging (QPI) and machine learning to model the health of RBCs in people with SCD. The health index combines assessment of cell deformation, sickle-shaped classification, and membrane flexibility to evaluate erythrocyte health. Using QPI and image processing, the percentage of sickle-shaped cells and membrane flexibility were quantified. Statistically significant differences were observed between individuals with and without SCD, indicating the impact of underlying pathophysiology on erythrocyte health. Additionally, sodium metabisulfite led to an increase in sickle-shaped cells and a decrease in flexibility in the sickle cell blood samples. Based on these findings, two approaches were used to calculate the Sickle Erythrocyte Health Index: one using hand-crafted features and one using learned features from deep learning models. Both indices showed significant differences between non-SCD and SCD groups and sensitivity to changes induced by sodium metabisulfite. The Sickle Erythrocyte Health Index has important clinical implications for SCD management and could be used by providers when making treatment decisions. Further research is warranted to evaluate the clinical utility and applicability of the Sickle Erythrocyte Health Index in diverse patient populations.
Collapse
Affiliation(s)
- Yaw Ofosu Nyansa Ansong-Ansongton
- Department of Bioengineering, KovaDx, New Haven, CT; Department of Bioengineering, University of California Berkeley, Bioengineering, Berkeley, CA.
| | - Timothy D Adamson
- Department of Bioengineering, KovaDx, New Haven, CT; Department of Bioengineering, University of California Berkeley, Bioengineering, Berkeley, CA
| |
Collapse
|
28
|
Kashyap Y, Wang ZJ. Gut microbiota dysbiosis alters chronic pain behaviors in a humanized transgenic mouse model of sickle cell disease. Pain 2024; 165:423-439. [PMID: 37733476 PMCID: PMC10843763 DOI: 10.1097/j.pain.0000000000003034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Pain is the most common symptom experienced by patients with sickle cell disease (SCD) throughout their lives and is the main cause of hospitalization. Despite the progress that has been made towards understanding the disease pathophysiology, major gaps remain in the knowledge of SCD pain, the transition to chronic pain, and effective pain management. Recent evidence has demonstrated a vital role of gut microbiota in pathophysiological features of SCD. However, the role of gut microbiota in SCD pain is yet to be explored. We sought to evaluate the compositional differences in the gut microbiota of transgenic mice with SCD and nonsickle control mice and investigate the role of gut microbiota in SCD pain by using antibiotic-mediated gut microbiota depletion and fecal material transplantation (FMT). The antibiotic-mediated gut microbiota depletion did not affect evoked pain but significantly attenuated ongoing spontaneous pain in mice with SCD. Fecal material transplantation from mice with SCD to wild-type mice resulted in tactile allodynia (0.95 ± 0.17 g vs 0.08 ± 0.02 g, von Frey test, P < 0.001), heat hyperalgesia (15.10 ± 0.79 seconds vs 8.68 ± 1.17 seconds, radiant heat, P < 0.01), cold allodynia (2.75 ± 0.26 seconds vs 1.68 ± 0.08 seconds, dry ice test, P < 0.01), and anxiety-like behaviors (Elevated Plus Maze Test, Open Field Test). On the contrary, reshaping gut microbiota of mice with SCD with FMT from WT mice resulted in reduced tactile allodynia (0.05 ± 0.01 g vs 0.25 ± 0.03 g, P < 0.001), heat hyperalgesia (5.89 ± 0.67 seconds vs 12.25 ± 0.76 seconds, P < 0.001), and anxiety-like behaviors. These findings provide insights into the relationship between gut microbiota dysbiosis and pain in SCD, highlighting the importance of gut microbial communities that may serve as potential targets for novel pain interventions.
Collapse
Affiliation(s)
- Yavnika Kashyap
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
| | - Zaijie Jim Wang
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
- Department of Neurology & Rehabilitation, and Sickle Cell Center, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, United States
| |
Collapse
|
29
|
Li W, Pucka AQ, Debats C, Reyes BA, Syed F, O’Brien AR, Mehta R, Manchanda N, Jacob SA, Hardesty BM, Greist A, Harte SE, Harris RE, Yu Q, Wang Y. Inflammation and autoimmunity are interrelated in patients with sickle cell disease at a steady-state condition: implications for vaso-occlusive crisis, pain, and sensory sensitivity. Front Immunol 2024; 15:1288187. [PMID: 38361924 PMCID: PMC10867278 DOI: 10.3389/fimmu.2024.1288187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
This study aimed to comprehensively analyze inflammatory and autoimmune characteristics of patients with sickle cell disease (SCD) at a steady-state condition (StSt) compared to healthy controls (HCs) to explore the pathogenesis of StSt and its impact on patients' well-being. The study cohort consisted of 40 StSt participants and 23 HCs enrolled between July 2021 and April 2023. StSt participants showed elevated white blood cell (WBC) counts and altered hematological measurements when compared to HCs. A multiplex immunoassay was used to profile 80 inflammatory cytokines/chemokines/growth factors in plasma samples from these SCD participants and HCs. Significantly higher plasma levels of 35 analytes were observed in SCD participants, with HGF, IL-18, IP-10, and MCP-2 being among the most significantly affected analytes. Additionally, autoantibody profiles were also altered, with elevated levels of anti-SSA/Ro60, anti-Ribosomal P, anti-Myeloperoxidase (MPO), and anti-PM/Scl-100 observed in SCD participants. Flow cytometric analysis revealed higher rates of red blood cell (RBC)/reticulocyte-leukocyte aggregation in SCD participants, predominantly involving monocytes. Notably, correlation analysis identified associations between inflammatory mediator levels, autoantibodies, RBC/reticulocyte-leukocyte aggregation, clinical lab test results, and pain crisis/sensitivity, shedding light on the intricate interactions between these factors. The findings underscore the potential significance of specific biomarkers and therapeutic targets that may hold promise for future investigations and clinical interventions tailored to the unique challenges posed by SCD. In addition, the correlations between vaso-occlusive crisis (VOC)/pain/sensory sensitivity and inflammation/immune dysregulation offer valuable insights into the pathogenesis of SCD and may lead to more targeted and effective therapeutic strategies. Clinical Trial Registration ClinicalTrials.gov, Identifier: NCT05045820.
Collapse
Affiliation(s)
- Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew Q. Pucka
- Department of Anesthesia, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Candice Debats
- Department of Anesthesia, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brandon A. Reyes
- Department of Anesthesia, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fahim Syed
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew R.W. O’Brien
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rakesh Mehta
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Naveen Manchanda
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Seethal A. Jacob
- Children’s Health Services Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Anne Greist
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN, United States
| | - Steven E. Harte
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Richard E. Harris
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, United States
- Susan Samueli Integrative Health Institute, and Department of Anesthesiology and Perioperative Care, School of Medicine, University of California at Irvine, Irvine, CA, United States
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ying Wang
- Department of Anesthesia, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
30
|
DeVeaux SA, Vyshnya S, Propsom K, Gbotosho OT, Singh AS, Horning RZ, Sharma M, Jegga AG, Niu L, Botchwey EA, Hyacinth HI. Neuroinflammation underlies the development of social stress induced cognitive deficit in sickle cell disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577074. [PMID: 38328164 PMCID: PMC10849745 DOI: 10.1101/2024.01.24.577074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Cognitive deficit is a debilitating complication of SCD with multifactorial pathobiology. Here we show that neuroinflammation and dysregulation in lipidomics and transcriptomics profiles are major underlying mechanisms of social stress-induced cognitive deficit in SCD. Townes sickle cell (SS) mice and controls (AA) were exposed to social stress using the repeat social defeat (RSD) paradigm concurrently with or without treatment with minocycline. Mice were tested for cognitive deficit using novel object recognition (NOR) and fear conditioning (FC) tests. SS mice exposed to RSD without treatment had worse performance on cognitive tests compared to SS mice exposed to RSD with treatment or to AA controls, irrespective of their RSD or treatment disposition. Additionally, compared to SS mice exposed to RSD with treatment, SS mice exposed to RSD without treatment had significantly more cellular evidence of neuroinflammation coupled with a significant shift in the differentiation of neural progenitor cells towards astrogliogenesis. Additionally, brain tissue from SS mice exposed to RSD was significantly enriched for genes associated with blood-brain barrier dysfunction, neuron excitotoxicity, inflammation, and significant dysregulation in sphingolipids important to neuronal cell processes. We demonstrate in this study that neuroinflammation and lipid dysregulation are potential underlying mechanisms of social stress-related cognitive deficit in SS mice.
Collapse
Affiliation(s)
- S’Dravious A. DeVeaux
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, USA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, USA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Katherine Propsom
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Asem S. Singh
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robert Z. Horning
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mihika Sharma
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Liang Niu
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA, USA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
31
|
Rab MAE, Kanne CK, Boisson C, Bos J, van Oirschot BA, Houwing ME, Renoux C, Bartels M, Rijneveld AW, Nur E, Cnossen MH, Joly P, Nader E, Fort R, Connes P, van Wijk R, Sheehan VA, van Beers EJ. Oxygen gradient ektacytometry-derived biomarkers are associated with acute complications in sickle cell disease. Blood Adv 2024; 8:276-286. [PMID: 37976458 PMCID: PMC10824684 DOI: 10.1182/bloodadvances.2023011013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023] Open
Abstract
ABSTRACT We investigated the potential of the point of sickling (PoS; the pO2 tension at which red cells start to sickle), determined by oxygen gradient ektacytometry to serve as a biomarker associated with the incidence of acute sickle cell disease-related complications in 177 children and 50 adults. In the pediatric cohort, for every 10 mmHg increase in PoS reflecting a greater likelihood of sickling, the likelihood of an individual experiencing >1 type of acute complication increased; the adjusted odds ratio (aOR) was 1.65. For every 0.1 increase in minimum elongation index (EImin; reflecting improved red blood cell deformability at hypoxia), the aOR was 0.50. In the adult cohort, for every 10 mmHg increase in PoS, we found an aOR of 3.00, although this was not significant after correcting for multiple testing. There was a trend for an association between higher PoS and greater likelihood of vaso-occlusive episodes (VOEs; children aOR, 1.35; adults aOR, 2.22). In children, only EImin was associated with VOEs (aOR, 0.68). When data of both cohorts were pooled, significant associations with PoS and/or EImin were found for all acute complications, independently and when >1 type of acute complication was assessed. These findings indicate that oxygen gradient ektacytometry generates novel biomarkers and provides a rationale for further development of these biomarkers in the assessment of clinical severity, evaluation of novel therapies, and as surrogate clinical trial end points. These biomarkers may be useful in assessing efficacy of novel therapies like pyruvate kinase activators, voxelotor, and L-glutamine.
Collapse
Affiliation(s)
- Minke A. E. Rab
- Central Diagnostic Laboratory-Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Celeste K. Kanne
- Department of Pediatrics Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, GA
| | - Camille Boisson
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Jennifer Bos
- Central Diagnostic Laboratory-Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Brigitte A. van Oirschot
- Central Diagnostic Laboratory-Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maite E. Houwing
- Department of Pediatric Hematology and Oncology, Erasmus Medical Center Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Céline Renoux
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
- Laboratory of Biochemistry and Molecular Biology, UF Biochemistry of Red Blood Cell Diseases, Est Center of Biology and Pathology, Hospices Civils de Lyon, Lyon, France
| | - Marije Bartels
- Van Creveldkliniek, Divison of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anita W. Rijneveld
- Department of Hematology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Erfan Nur
- Department of Hematology, Amsterdam University Medical Center, The Netherlands
| | - Marjon H. Cnossen
- Department of Pediatric Hematology and Oncology, Erasmus Medical Center Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Philippe Joly
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
- Laboratory of Biochemistry and Molecular Biology, UF Biochemistry of Red Blood Cell Diseases, Est Center of Biology and Pathology, Hospices Civils de Lyon, Lyon, France
| | - Elie Nader
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Romain Fort
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
- Department of Internal Medicine, Hospices Civils de Lyon, Lyon, France
| | - Philippe Connes
- Laboratory LIBM EA7424, University of Lyon 1, “Vascular Biology and Red Blood Cell” team, Lyon, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Richard van Wijk
- Central Diagnostic Laboratory-Research, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Vivien A. Sheehan
- Department of Pediatrics Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, GA
| | - Eduard J. van Beers
- Van Creveldkliniek, Divison of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
32
|
Bell V, Varzakas T, Psaltopoulou T, Fernandes T. Sickle Cell Disease Update: New Treatments and Challenging Nutritional Interventions. Nutrients 2024; 16:258. [PMID: 38257151 PMCID: PMC10820494 DOI: 10.3390/nu16020258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Sickle cell disease (SCD), a distinctive and often overlooked illness in the 21st century, is a congenital blood disorder characterized by considerable phenotypic diversity. It comprises a group of disorders, with sickle cell anemia (SCA) being the most prevalent and serious genotype. Although there have been some systematic reviews of global data, worldwide statistics regarding SCD prevalence, morbidity, and mortality remain scarce. In developed countries with a lower number of sickle cell patients, cutting-edge technologies have led to the development of new treatments. However, in developing settings where sickle cell disease (SCD) is more prevalent, medical management, rather than a cure, still relies on the use of hydroxyurea, blood transfusions, and analgesics. This is a disease that affects red blood cells, consequently affecting most organs in diverse manners. We discuss its etiology and the advent of new technologies, but the aim of this study is to understand the various types of nutrition-related studies involving individuals suffering from SCD, particularly in Africa. The interplay of the environment, food, gut microbiota, along with their respective genomes collectively known as the gut microbiome, and host metabolism is responsible for mediating host metabolic phenotypes and modulating gut microbiota. In addition, it serves the purpose of providing essential nutrients. Moreover, it engages in direct interactions with host homeostasis and the immune system, as well as indirect interactions via metabolites. Nutrition interventions and nutritional care are mechanisms for addressing increased nutrient expenditures and are important aspects of supportive management for patients with SCD. Underprivileged areas in Sub-Saharan Africa should be accompanied by efforts to define and promote of the nutritional aspects of SCD. Their importance is key to maintaining well-being and quality of life, especially because new technologies and products remain limited, while the use of native medicinal plant resources is acknowledged.
Collapse
Affiliation(s)
- Victoria Bell
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Theodoros Varzakas
- Department of Food Science and Technology, University of the Peloponnese, 24100 Kalamata, Greece
| | - Theodora Psaltopoulou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Tito Fernandes
- CIISA, Faculty of Veterinary Medicine, University of Lisbon, 1649-004 Lisbon, Portugal
| |
Collapse
|
33
|
Obeagu EI, Ubosi NI, Obeagu GU, Egba SI, Bluth MH. Understanding apoptosis in sickle cell anemia patients: Mechanisms and implications. Medicine (Baltimore) 2024; 103:e36898. [PMID: 38215146 PMCID: PMC10783340 DOI: 10.1097/md.0000000000036898] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024] Open
Abstract
Sickle cell anemia (SCA) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin, leading to the formation of sickle-shaped red blood cells. While much research has focused on the molecular and cellular mechanisms underlying the pathophysiology of SCA, recent attention has turned to the role of apoptosis, or programmed cell death, in the disease progression. This review aims to elucidate the intricate mechanisms of apoptosis in SCA patients and explore its implications in disease severity, complications, and potential therapeutic interventions. Different research search engines such as PubMed central, Scopus, Web of Science, Google Scholar, ResearchGate, Academia Edu, etc were utilized in writing this paper. Apoptosis, a highly regulated cellular process, plays a crucial role in maintaining homeostasis by eliminating damaged or dysfunctional cells. In SCA, the imbalance between pro-apoptotic and anti-apoptotic signals contributes to increased erythrocyte apoptosis, exacerbating anemia and vaso-occlusive crises. Various factors, including oxidative stress, inflammation, and altered cell signaling pathways, converge to modulate the apoptotic response in SCA. Furthermore, the interaction between apoptotic cells and the vascular endothelium contributes to endothelial dysfunction, promoting the pathogenesis of vasculopathy and organ damage seen in SCA patients. In conclusion, unraveling the complexities of apoptosis in SCA provides valuable insights into the disease pathophysiology and offers novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
| | - Nwanganga Ihuoma Ubosi
- Department of Public Health Sciences, Faculty of Health Sciences, National Open University of Nigeria, Headquarters, Jabi, Abuja, Nigeria
| | | | - Simeon Ikechukwu Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | - Martin H. Bluth
- Department of Pathology, Division of Blood Transfusion Medicine, Maimonides Medical Center, Brooklyn, NY, USA
| |
Collapse
|
34
|
Smiianov VV, Fartushok TV, Fedevych YM, Fartushok NV. New view on the compatibility of hemoglobin function in the erythrocytes. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:9-16. [PMID: 38431801 DOI: 10.36740/wlek202401101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To study the process of hemoglobin oxidation and the enzymatic reactions associated with it. PATIENTS AND METHODS Materials and Methods: Heparinized human blood (15 IU/ml) was obtained from the clinical department. The concentration of oxy- and methemoglobin, auto-oxidation of hemoglobin was determined spectrophotometrically spectrophotometrically. Autooxidation of hemoglobin was recorded spectrophotometrically, and protein concentration was determined by the Lowry method. Monooxygenase activity of hemoglobin was also measured by the method described by Lowry spectrophotometrically. The concentration of O2 and H2O2 in the reaction media was determined on a biomicroanalyzer OR 210/3 (Redelkis). RESULTS Results: The obtained experimental data allow us to propose a mechanism of "spontaneous autooxidation" of oxyhemoglobin, which can be described by the following equations: Hb2+O2 → Hb3+ + O2 - (1) Hb2+O2 + 2e - + 2H+ →Hb3+ + H2O2 (2) Hb2+O2 + 2e - + 2H+ →Hb2+ + H2O2 (3) Hb2+ + O2 →Hb2+O2 (4) Spectral characteristics of the process of "spontaneous auto-oxidation" indicate the formation of a metform of hemoglobin, the depletion of oxygen by the system was established, at pH 5.6, an increase in the monooxygenase activity of hemoglobin is observed 3-4 times compared to the physiological level. CONCLUSION Сonclusions: In addition to the main, previously known functions of hemoglobin (gas transport, peroxidase, monooxygenase), it catalyzes a two-electron oxidase reaction in which O2 is reduced to H2O2. This is confirmed by experimental data on the formation of one of the products of "spontaneous autoxidation" of oxyhemoglobin _ deoxyform at pH 5.6 _ 8.9.
Collapse
Affiliation(s)
| | | | - Yuri M Fedevych
- DANYLO HALYTSKY LVIV NATIONAL MEDICAL UNIVERSITY, LVIV UKRAINE
| | | |
Collapse
|
35
|
Connes P. Blood rheology and vascular function in sickle cell trait and sickle cell disease: From pathophysiological mechanisms to clinical usefulness. Clin Hemorheol Microcirc 2024; 86:9-27. [PMID: 38073384 DOI: 10.3233/ch-238122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Sickle cell disease (SCD) is an autosomal recessive disorder. Although the molecular mechanisms at the origin of SCD have been well characterized, its clinical expression is highly variable. SCD is characterized by blood rheological abnormalities, increased inflammation and oxidative stress, and vascular dysfunction. Individuals with only one copy of the mutated β-globin gene have sickle cell trait (SCT) and are usually asymptomatic. The first part of this review focuses on the biological responses of SCT carriers during exercise and on the effects of combined SCT and diabetes on vascular function, several biomarkers and clinical complications. The second part of the review focuses on SCD and shows that the magnitude of red blood cell (RBC) rheological alterations is highly variable from one patient to another, and this variability reflects the clinical and hematological variability: patients with the less deformable RBCs have high hemolytic rate and severe anemia, and are prone to develop leg ulcers, priapism, cerebral vasculopathy, glomerulopathy or pulmonary hypertension. In contrast, SCD patients characterized by the presence of more deformable RBCs (but still rigid) are less anemic and may exhibit increased blood viscosity, which increases the risk for vaso-occlusive events. Several genetic and cellular factors may modulate RBC deformability in SCD: co-existence of α-thalassemia, fetal hemoglobin level, oxidative stress, the presence of residual mitochondria into mature RBCs, the activity of various non-selective cationic ion channels, etc. The last part of this review presents the effects of hydroxyurea and exercise training on RBC rheology and other biomarkers in SCD.
Collapse
Affiliation(s)
- Philippe Connes
- Laboratory LIBM EA7424, University of Lyon 1, "Vascular Biology and Red Blood Cell" Team, Lyon, France
- Laboratory of Excellence Labex GR-Ex, Paris, France
| |
Collapse
|
36
|
Connes P, Renoux C, Joly P, Nader E. Vascular pathophysiology of sickle cell disease. Presse Med 2023; 52:104202. [PMID: 37944640 DOI: 10.1016/j.lpm.2023.104202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Sickle cell disease (SCD) is an hereditary disorder characterized by the production of an abnormal hemoglobin called hemoglobin S (HbS). HbS may polymerize in deoxygenated conditions, which leads to red blood cell (RBC) sickling. Sickled RBCs are more rigid and fragile, and prone to lysis. SCD patients exhibit various acute and/or chronic complications, which may affect several organs. The clinical presentation of SCD is highly variable from one patient to another and cannot be only explained by RBC sickling. Increased blood viscosity, caused by the presence of RBCs with abnormal deformability and aggregation, may increase vascular resistance and increase the risk of acute and chronic vascular complications. Chronic hemolysis results in decreased nitric oxide (NO) bioavailability which may compromise vasodilation and participate to the development of chronic vasculopathy. Furthermore, chronic hemolysis is responsible for increased inflammation and oxidative stress, which affect the vascular system and may promote the adhesion of circulating cells to endothelial cells. Extracellular vesicles and especially RBC microparticles (massively released in the context of SCD) are also at the origin of vascular damages and increased white blood cells adhesion to the endothelium, which may trigger vaso-occlusive crisis and other vascular-related complications. This review highlights the fact that SCD should not only be considered as a hematological disorder but also as a vascular disease.
Collapse
Affiliation(s)
- 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, France; Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.
| | - Céline Renoux
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, France; Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France; Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - Philippe Joly
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, France; Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France; Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi-site, Hospices Civils de Lyon, Lyon, France
| | - 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, France; Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| |
Collapse
|
37
|
Nathan DI, Dougherty M, Bhatta M, Mascarenhas J, Marcellino BK. Clonal hematopoiesis and inflammation: A review of mechanisms and clinical implications. Crit Rev Oncol Hematol 2023; 192:104187. [PMID: 37879493 DOI: 10.1016/j.critrevonc.2023.104187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/21/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Clonal hematopoiesis (CH) is defined by the presence of somatic mutations in hematopoietic stem and progenitor cells (HSPC). CH is associated primarily with advancing age and confers an elevated risk of progression to overt hematologic malignancy and cardiovascular disease. Increasingly, CH is associated with a wide range of diseases driven by, and sequelae of, inflammation. Accordingly, there is great interest in better understanding the pathophysiologic and clinical relationship between CH, aging, and disease. Both observational and experimental findings support the concept that CH is a potential common denominator in the inflammatory outcomes of aging. However, there is also evidence that local and systemic inflammatory states promote the growth and select for CH clones. In this review, we aim to provide an up-to-date summary of the nature of the relationship between inflammation and CH, which is central to unlocking potential therapeutic opportunities to prevent progression to myeloid malignancy.
Collapse
Affiliation(s)
- Daniel I Nathan
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Max Dougherty
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manasa Bhatta
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bridget K Marcellino
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Williams DC, Wood DK. High-throughput quantification of red blood cell deformability and oxygen saturation to probe mechanisms of sickle cell disease. Proc Natl Acad Sci U S A 2023; 120:e2313755120. [PMID: 37983504 PMCID: PMC10691249 DOI: 10.1073/pnas.2313755120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023] Open
Abstract
The complex, systemic pathology of sickle cell disease is driven by multiple mechanisms including red blood cells (RBCs) stiffened by polymerized fibers of deoxygenated sickle hemoglobin. A critical step toward understanding the pathologic role of polymer-containing RBCs is quantifying the biophysical changes in these cells in physiologically relevant oxygen environments. We have developed a microfluidic platform capable of simultaneously measuring single RBC deformability and oxygen saturation under controlled oxygen and shear stress. We found that RBCs with detectable amounts of polymer have decreased oxygen affinity and decreased deformability. Surprisingly, the deformability of the polymer-containing cells is oxygen-independent, while the fraction of these cells increases as oxygen decreases. We also find that some fraction of these cells is present at most physiologic oxygen tensions, suggesting a role for these cells in the systemic pathologies. Additionally, the ability to measure these pathological cells should provide clearer targets for evaluating therapies.
Collapse
Affiliation(s)
- Dillon C. Williams
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN55455
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
39
|
Alibayov B, Scasny A, Vidal AGJ, Murin L, Wong S, Edwards KS, Eichembaun Z, Punshon T, Jackson BP, Hopp MT, McDaniel LS, Akerley BJ, Imhof D, Vidal JE. Oxidation of hemoglobin in the lung parenchyma facilitates the differentiation of pneumococci into encapsulated bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567109. [PMID: 38014009 PMCID: PMC10680745 DOI: 10.1101/2023.11.14.567109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Pneumococcal pneumonia causes cytotoxicity in the lung parenchyma but the underlying mechanism involves multiple factors contributing to cell death. Here, we discovered that hydrogen peroxide produced by Streptococcus pneumoniae (Spn-H 2 O 2 ) plays a pivotal role by oxidizing hemoglobin, leading to its polymerization and subsequent release of labile heme. At physiologically relevant levels, heme selected a population of encapsulated pneumococci. In the absence of capsule and Spn-H 2 O 2 , host intracellular heme exhibited toxicity towards pneumococci, thus acting as an antibacterial mechanism. Further investigation revealed that heme-mediated toxicity required the ABC transporter GlnPQ. In vivo experiments demonstrated that pneumococci release H 2 O 2 to cause cytotoxicity in bronchi and alveoli through the non-proteolytic degradation of intracellular proteins such as actin, tubulin and GAPDH. Overall, our findings uncover a mechanism of lung toxicity mediated by oxidative stress that favor the growth of encapsulated pneumococci suggesting a therapeutic potential by targeting oxidative reactions. Graphical abstract Highlights Oxidation of hemoglobin by Streptococcus pneumoniae facilitates differentiation to encapsulated pneumococci in vivo Differentiated S. pneumoniae produces capsule and hydrogen peroxide (Spn-H 2 O 2 ) as defense mechanism against host heme-mediated toxicity. Spn-H 2 O 2 -induced lung toxicity causes the oxidation and non-proteolytic degradation of intracellular proteins tubulin, actin, and GAPDH. The ABC transporter GlnPQ is a heme-binding complex that makes Spn susceptible to heme toxicity.
Collapse
|
40
|
Esperti S, Nader E, Stier A, Boisson C, Carin R, Marano M, Robert M, Martin M, Horand F, Cibiel A, Renoux C, Van Bruggen R, Blans C, Dargaud Y, Joly P, Gauthier A, Poutrel S, Romana M, Roussel D, Connes P. Increased retention of functional mitochondria in mature sickle red blood cells is associated with increased sickling tendency, hemolysis and oxidative stress. Haematologica 2023; 108:3086-3094. [PMID: 37259576 PMCID: PMC10620576 DOI: 10.3324/haematol.2023.282684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Abnormal retention of mitochondria in mature red blood cells (RBC) has been recently reported in sickle cell anemia (SCA) but their functionality and their role in the pathophysiology of SCA remain unknown. The presence of mitochondria within RBC was determined by flow cytometry in 61 SCA patients and ten healthy donors. Patients were classified according to the percentage of mature RBC with mitochondria contained in the whole RBC population: low (0-4%), moderate (>4% and <8%), or high level (>8%). RBC rheological, hematological, senescence and oxidative stress markers were compared between the three groups. RBC senescence and oxidative stress markers were also compared between mature RBC containing mitochondria and those without. The functionality of residual mitochondria in sickle RBC was measured by high-resolution respirometry assay and showed detectable mitochondrial oxygen consumption in sickle mature RBC but not in healthy RBC. Increased levels of mitochondrial reactive oxygen species were observed in mature sickle RBC when incubated with Antimycin A versus without. In addition, mature RBC retaining mitochondria exhibited greater levels of reactive oxygen species compared to RBC without mitochondria, as well as greater Ca2+, lower CD47 and greater phosphatidylserine exposure. Hematocrit and RBC deformability were lower, and the propensity of RBC to sickle under deoxygenation was higher, in the SCA group with a high percentage of mitochondria retention in mature RBC. This study showed the presence of functional mitochondria in mature sickle RBC, which could favor RBC sickling and accelerate RBC senescence, leading to increased cellular fragility and hemolysis.
Collapse
Affiliation(s)
- Sofia Esperti
- 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, 79015 Paris, France; Erytech Pharma, 69008 Lyon
| | - 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, 79015 Paris
| | - Antoine Stier
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés, CNRS, ENTPE, UMR 5023 Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France; Université de Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR7178, 67000 Strasbourg
| | - Camille Boisson
- 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, 79015 Paris
| | - Romain Carin
- 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, 79015 Paris
| | - Muriel Marano
- UR4609 Hémostase and Thrombose Université Claude Bernard Lyon 1, Université de Lyon, Lyon
| | - Mélanie Robert
- 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, 79015 Paris, France; Erytech Pharma, 69008 Lyon
| | - Marie Martin
- 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
| | | | | | - Céline Renoux
- 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, 79015 Paris, France; Laboratoire de Biochimie et de Biologie Moléculaire, UF de Biochimie des Pathologies Erythrocytaires, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 69500 Lyon
| | - Robin Van Bruggen
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, The Netherlands
| | - Colin Blans
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, The Netherlands
| | - Yesim Dargaud
- UR4609 Hémostase and Thrombose Université Claude Bernard Lyon 1, Université de Lyon, Lyon
| | - Philippe Joly
- 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, 79015 Paris, France; Laboratoire de Biochimie et de Biologie Moléculaire, UF de Biochimie des Pathologies Erythrocytaires, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 69500 Lyon
| | - Alexandra Gauthier
- 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, 79015 Paris, France; Institut d'Hématologique et d'Oncologique Pédiatrique, Hospices Civils de Lyon, 69008 Lyon
| | - Solène Poutrel
- 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, 79015 Paris, France; Service de Médecine Interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, 69008 Lyon, France. de Médecine Interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, 69008 Lyon
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, 79015 Paris, France; Université de Paris, Université des Antilles, UMR_S1134, BIGR, INSERM, Paris
| | - Damien Roussel
- Laboratoire d'Ecologie des Hydrosystèmes Naturels et Anthropisés, CNRS, ENTPE, UMR 5023 Université Claude Bernard Lyon 1, Université de Lyon, Lyon
| | - 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, 79015 Paris.
| |
Collapse
|
41
|
Li W, Pucka AQ, Debats C, Reyes B, Syed F, O'Brien AR, Mehta R, Manchanda N, Jacob SA, Hardesty BM, Greist A, Harte SE, Harris RE, Yu Q, Wang Y. Inflammation and autoimmunity are interrelated in patients with sickle cell disease at a steady-state condition: implications for vaso-occlusive crisis, pain, and sensory sensitivity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.03.23294996. [PMID: 37732268 PMCID: PMC10508800 DOI: 10.1101/2023.09.03.23294996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
This study aimed to comprehensively analyze inflammatory and autoimmune characteristics of patients with sickle cell disease (SCD) at a steady-state condition (StSt) compared to healthy controls (HCs) to explore the pathogenesis of StSt and its impact on patients' well-being. The study cohort consisted of 40 StSt participants and 23 HCs enrolled between July 2021 and April 2023. StSt participants showed elevated white blood cell (WBC) counts and altered hematological measurements when compared to HCs. A multiplex immunoassay was used to profile 80 inflammatory cytokines/chemokines/growth factors in plasma samples from these SCD participants and HCs. Significantly higher plasma levels of 37 analytes were observed in SCD participants, with HGF, IL-18, IP-10, and MCP-2 being among the most significantly affected analytes. Additionally, autoantibody profiles were also altered, with elevated levels of anti-SSA/Ro60, anti-Ribosomal P, anti-Myeloperoxidase (MPO), and anti-PM/Scl-100 observed in SCD participants. Flow cytometric analysis revealed higher rates of red blood cell (RBC)/reticulocyte-leukocyte aggregation in SCD participants, predominantly involving monocytes. Notably, correlation analysis identified associations between inflammatory mediator levels, autoantibodies, RBC/reticulocyte-leukocyte aggregation, clinical lab test results, and pain crisis/sensitivity, shedding light on the intricate interactions between these factors. The findings underscore the potential significance of specific biomarkers and therapeutic targets that may hold promise for future investigations and clinical interventions tailored to the unique challenges posed by SCD. In addition, the correlations between vaso-occlusive crisis (VOC)/pain/sensory sensitivity and inflammation/immune dysregulation offer valuable insights into the pathogenesis of SCD and may lead to more targeted and effective therapeutic strategies.
Collapse
|
42
|
Aboderin FI, Oduola T, Davison GM, Oguntibeju OO. A Review of the Relationship between the Immune Response, Inflammation, Oxidative Stress, and the Pathogenesis of Sickle Cell Anaemia. Biomedicines 2023; 11:2413. [PMID: 37760854 PMCID: PMC10525295 DOI: 10.3390/biomedicines11092413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Sickle cell anaemia (SCD) is a life-threatening haematological disorder which is predominant in sub-Saharan Africa and is triggered by a genetic mutation of the β-chain haemoglobin gene resulting in the substitution of glutamic acid with valine. This mutation leads to the production of an abnormal haemoglobin molecule called haemoglobin S (HbS). When deoxygenated, haemoglobin S (HbS) polymerises and results in a sickle-shaped red blood cell which is rigid and has a significantly shortened life span. Various reports have shown a strong link between oxidative stress, inflammation, the immune response, and the pathogenesis of sickle cell disease. The consequence of these processes leads to the development of vasculopathy (disease of the blood vessels) and several other complications. The role of the immune system, particularly the innate immune system, in the pathogenesis of SCD has become increasingly clear in recent years of research; however, little is known about the roles of the adaptive immune system in this disease. This review examines the interaction between the immune system, inflammation, oxidative stress, blood transfusion, and their effects on the pathogenesis of sickle cell anaemia.
Collapse
Affiliation(s)
- Florence Ifechukwude Aboderin
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Taofeeq Oduola
- Department of Chemical Pathology, Usmanu Danfodiyo University, Sokoto 840004, Nigeria;
| | - Glenda Mary Davison
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Oluwafemi Omoniyi Oguntibeju
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| |
Collapse
|
43
|
Fay ME, Oshinowo O, Iffrig E, Fibben KS, Caruso C, Hansen S, Musick JO, Valdez JM, Azer SS, Mannino RG, Choi H, Zhang DY, Williams EK, Evans EN, Kanne CK, Kemp ML, Sheehan VA, Carden MA, Bennett CM, Wood DK, Lam WA. iCLOTS: open-source, artificial intelligence-enabled software for analyses of blood cells in microfluidic and microscopy-based assays. Nat Commun 2023; 14:5022. [PMID: 37596311 PMCID: PMC10439163 DOI: 10.1038/s41467-023-40522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
While microscopy-based cellular assays, including microfluidics, have significantly advanced over the last several decades, there has not been concurrent development of widely-accessible techniques to analyze time-dependent microscopy data incorporating phenomena such as fluid flow and dynamic cell adhesion. As such, experimentalists typically rely on error-prone and time-consuming manual analysis, resulting in lost resolution and missed opportunities for innovative metrics. We present a user-adaptable toolkit packaged into the open-source, standalone Interactive Cellular assay Labeled Observation and Tracking Software (iCLOTS). We benchmark cell adhesion, single-cell tracking, velocity profile, and multiscale microfluidic-centric applications with blood samples, the prototypical biofluid specimen. Moreover, machine learning algorithms characterize previously imperceptible data groupings from numerical outputs. Free to download/use, iCLOTS addresses a need for a field stymied by a lack of analytical tools for innovative, physiologically-relevant assays of any design, democratizing use of well-validated algorithms for all end-user biomedical researchers who would benefit from advanced computational methods.
Collapse
Affiliation(s)
- Meredith E Fay
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elizabeth Iffrig
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Kirby S Fibben
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christina Caruso
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Scott Hansen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Jamie O Musick
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - José M Valdez
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sally S Azer
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert G Mannino
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyoann Choi
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Dan Y Zhang
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Evelyn K Williams
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Erica N Evans
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Celeste K Kanne
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Vivien A Sheehan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Marcus A Carden
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Carolyn M Bennett
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
44
|
Aldali JA, Alotaibi BA, Aldali HJ, Alasiri GA, Alaseem A, Almuqrin AM, Alshalani A, Alotaibi FT. Assessing the Impact of COVID-19 Vaccines on Sickle Cell Anaemia Patients: A Comparative Analysis of Biochemical and Haematological Parameters. Biomedicines 2023; 11:2203. [PMID: 37626700 PMCID: PMC10452730 DOI: 10.3390/biomedicines11082203] [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: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) vaccines have been developed to help prevent the spread of the virus infections. The COVID-19 vaccines, including Pfizer, Moderna, and AstraZeneca, have undergone rigorous testing and have demonstrated both safety and effectiveness. Extensive evidence supports their effectiveness in preventing severe illness, hospitalization, and mortality associated with COVID-19 infection. The administration of COVID-19 vaccines can directly affect hematological and biochemical parameters, with reported cases showing an association with thrombosis and thrombocytopenia. Therefore, it was hypothesized that COVID-19 vaccines may also influence hematological and biochemical markers in sickle cell patients. This study aimed to investigate the side effects of COVID-19 vaccines on sickle cell patients, providing a comprehensive evaluation of hematological and biochemical parameters. To our knowledge, this is the first study of its kind conducted in Saudi Arabia. The study included the evaluation of Pfizer and Oxford-AstraZeneca vaccines in sickle cell patients, measuring key parameters. Our findings revealed varying impacts of both vaccines on the ALT, AST, and CRP levels. Notably, CRP and ALT exhibited potential as indicators for renal disease, diabetes, and arthritis. However, further investigations are necessary to uncover the underlying mechanisms that drive these observed differences and comprehend their clinical implications for this vulnerable patient population. The unique nature of our study fills a crucial research gap and underscores the need for additional research in this area.
Collapse
Affiliation(s)
- Jehad A. Aldali
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Badi A. Alotaibi
- Department of Clinical Laboratory Sciences, Collage of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Hamzah J. Aldali
- Cellular and Molecular Medicine, College of Biomedical Science, University of Bristol, Bristol BS8 1QU, UK
| | - Glowi A. Alasiri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Ali Alaseem
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Abdulaziz M. Almuqrin
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Abdulrahman Alshalani
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Fahad T. Alotaibi
- Department of Physiology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 5701, Saudi Arabia
| |
Collapse
|
45
|
Kriebel J, Gonçalves IM, Baptista V, Veiga MI, Minas G, Lima R, Catarino SO. Extensional flow for assessing the effect of nanocarriers on the mechanical deformability of red blood cells. EXPERIMENTAL THERMAL AND FLUID SCIENCE 2023; 146:110931. [DOI: 10.1016/j.expthermflusci.2023.110931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
Seo J, Kim Y, Ji S, Kim HB, Jung H, Yi EC, Lee YH, Shin I, Yang WH, Cho JW. O-GlcNAcylation of RIPK1 rescues red blood cells from necroptosis. Front Immunol 2023; 14:1160490. [PMID: 37359541 PMCID: PMC10289004 DOI: 10.3389/fimmu.2023.1160490] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Necroptosis is a type of cell death with excessive inflammation and organ damage in various human diseases. Although abnormal necroptosis is common in patients with neurodegenerative, cardiovascular, and infectious diseases, the mechanisms by which O-GlcNAcylation contributes to the regulation of necroptotic cell death are poorly understood. In this study, we reveal that O-GlcNAcylation of RIPK1 (receptor-interacting protein kinase1) was decreased in erythrocytes of the mouse injected with lipopolysaccharide, resulting in the acceleration of erythrocyte necroptosis through increased formation of RIPK1-RIPK3 complex. Mechanistically, we discovered that O-GlcNAcylation of RIPK1 at serine 331 in human (corresponding to serine 332 in mouse) inhibits phosphorylation of RIPK1 at serine 166, which is necessary for the necroptotic activity of RIPK1 and suppresses the formation of the RIPK1-RIPK3 complex in Ripk1 -/- MEFs. Thus, our study demonstrates that RIPK1 O-GlcNAcylation serves as a checkpoint to suppress necroptotic signaling in erythrocytes.
Collapse
Affiliation(s)
- Junghwa Seo
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
| | - Yeolhoe Kim
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Suena Ji
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
| | - Han Byeol Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeryeon Jung
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eugene C. Yi
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yong-ho Lee
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Injae Shin
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Won Ho Yang
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jin Won Cho
- Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
47
|
Antonelli Rossi DA, De Araujo Junior JA, Luvizutto GJ, Bazan R, Salmazo PS, Modolo GP, Hueb JC, Nunes HRDC, Hokama NK, Minicucci MF, Roscani MG, Zanati Bazan SG. Effect of a Physical Exercise Program on the Inflammatory Response, Cardiac Functions, Functional Capacity, and Quality of Life in Patients with Sickle Cell Disease. J Clin Med 2023; 12:3952. [PMID: 37373647 PMCID: PMC10299033 DOI: 10.3390/jcm12123952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/13/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION The beneficial effects of physical exercise on functional capacity and inflammatory response are well-known in cardiovascular diseases; however, studies on sickle cell disease (SCD) are limited. It was hypothesized that physical exercise may exert a favorable effect on the inflammatory response of SCD patients, contributing to an improved quality of life. This study aimed to evaluate the effect of a regular physical exercise program on the anti-inflammatory responses in SCD patients. METHODS A non-randomized clinical trial was conducted in adult SCD patients. The patients were divided into two groups: 1-Exercise Group, which received a physical exercise program three times a week for 8 weeks, and; 2-Control Group, with routine physical activities. All patients underwent the following procedures initially and after eight weeks of protocol: clinical evaluation, physical evaluation, laboratory evaluation, quality of life evaluation, and echocardiographic evaluation. STATISTICAL ANALYSIS Comparisons between groups were made using Student's t-test, Mann-Whitney test, chi-square test, or Fisher's exact test. Spearman's correlation coefficient was calculated. The significance level was set at p < 0.05. RESULTS There was no significant difference in inflammatory response between the Control and Exercise Groups. The Exercise Group showed an improvement in peak VO2 values (p < 0.001), an increase in the distance walked (p < 0.001), an improvement in the limitation domain due to the physical aspects of the 36-Item Short Form Health Survey (SF-36) quality of life questionnaire (p = 0.022), and an increase in physical activity related to leisure (p < 0.001) and walking (p = 0.024) in the International Physical Activity Questionnaire (IPAQ). There was a negative correlation between IL-6 values and distance walked on the treadmill (correlation coefficient -0.444, p = 0.020) and the estimated peak VO2 values (correlation coefficient -0.480; p = 0.013) in SCD patients in both groups. CONCLUSIONS The aerobic exercise program did not change the inflammatory response profile of SCD patients, nor did it show unfavorable effects on the parameters evaluated, and patients with lower functional capacity were those with the highest levels of IL-6.
Collapse
Affiliation(s)
- Daniele Andreza Antonelli Rossi
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Jonas Alves De Araujo Junior
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Gustavo José Luvizutto
- Department of Applied Physical Therapy, Federal University of Triângulo Mineiro, Uberaba 38065-430, Brazil;
| | - Rodrigo Bazan
- Department of Neurology, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-970, Brazil; (R.B.); (G.P.M.)
| | - Péricles Sidnei Salmazo
- Department of Medicine, Faculty of Medical and Health Sciences, Pontifical Catholic University of São Paulo, Sorocaba 18052-490, Brazil;
| | - Gabriel Pinheiro Modolo
- Department of Neurology, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-970, Brazil; (R.B.); (G.P.M.)
| | - João Carlos Hueb
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | | | - Newton Key Hokama
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Marcos Ferreira Minicucci
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| | - Meliza Goi Roscani
- Department of Medicine, Federal University of Sao Carlos-UFSCar, São Carlos 13565-251, Brazil;
| | - Silméia Garcia Zanati Bazan
- Department of Internal Medicine, Botucatu Medical School-UNESP, São Paulo State University, Botucatu 18618-687, Brazil; (D.A.A.R.); (J.A.D.A.J.); (J.C.H.); (N.K.H.); (M.F.M.)
| |
Collapse
|
48
|
Nasir NJM, Heemskerk H, Jenkins J, Hamadee NH, Bunte R, Tucker-Kellogg L. Myoglobin-derived iron causes wound enlargement and impaired regeneration in pressure injuries of muscle. eLife 2023; 12:85633. [PMID: 37267120 DOI: 10.7554/elife.85633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 06/04/2023] Open
Abstract
The reasons for poor healing of pressure injuries are poorly understood. Vascular ulcers are worsened by extracellular release of hemoglobin, so we examined the impact of myoglobin (Mb) iron in murine muscle pressure injuries (mPI). Tests used Mb-knockout or treatment with deferoxamine iron chelator (DFO). Unlike acute injuries from cardiotoxin, mPI regenerated poorly with a lack of viable immune cells, persistence of dead tissue (necro-slough), and abnormal deposition of iron. However, Mb-knockout or DFO-treated mPI displayed a reversal of the pathology: decreased tissue death, decreased iron deposition, decrease in markers of oxidative damage, and higher numbers of intact immune cells. Subsequently, DFO treatment improved myofiber regeneration and morphology. We conclude that myoglobin iron contributes to tissue death in mPI. Remarkably, a large fraction of muscle death in untreated mPI occurred later than, and was preventable by, DFO treatment, even though treatment started 12 hr after pressure was removed. This demonstrates an opportunity for post-pressure prevention to salvage tissue viability.
Collapse
Affiliation(s)
- Nurul Jannah Mohamed Nasir
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Hans Heemskerk
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| | - Julia Jenkins
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Ralph Bunte
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Lisa Tucker-Kellogg
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| |
Collapse
|
49
|
Veshkini A, Gnott M, Vogel L, Kröger-Koch C, Tuchscherer A, Tröscher A, Bernabucci U, Trevisi E, Starke A, Mielenz M, Bachmann L, Hammon HM. Abomasal infusion of essential fatty acids and conjugated linoleic acid during late pregnancy and early lactation affects immunohematological and oxidative stress markers in dairy cows. J Dairy Sci 2023:S0022-0302(23)00231-X. [PMID: 37173257 DOI: 10.3168/jds.2022-22514] [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: 07/12/2022] [Accepted: 01/05/2023] [Indexed: 05/15/2023]
Abstract
Oxidative stress and inflammation, as natural parts of metabolic adaptations during the transition from late gestation to early lactation, are critical indicators of dairy cows' metabolic health. This study was designed to investigate the effects of abomasal infusion of essential fatty acids (EFA), particularly α-linolenic acid, and conjugated linoleic acid (CLA) on plasma, erythrocyte, and liver markers of oxidative stress in dairy cows during the transition period. Rumen-cannulated German Holstein cows (n = 38) in their second lactation (11,101 ± 1,118 kg milk/305 d, mean ± standard deviation) were abomasally infused with one of the following treatments from d -63 antepartum until d 63 postpartum (PP): CTRL (n = 9; 76 g/d coconut oil); EFA (n = 9; 78 g/d linseed plus 4 g/d safflower oil); CLA (n = 10; isomers cis-9,trans-11 and trans-10,cis-12 CLA; 38 g/d); and EFA+CLA (n = 10; 120 g/d). Hematological parameters as well as markers of oxidative status were measured in plasma, erythrocytes, and liver before and after calving. Immunohematological parameters, including erythrocyte number, hematocrit, hemoglobin, mean corpuscular hemoglobin, leukocytes, and basophils, were affected by time, and their peak levels were observed on the day after calving. The oxidative stress markers glutathione peroxidase 1 and reactive oxygen metabolites in plasma and erythrocytes were both affected by time, exhibiting the highest levels on d 1 PP, whereas β-carotene, retinol, and tocopherol were at their lowest levels at the same time. Immunohematological parameters were only marginally affected by fatty acid treatment in a time-dependent manner. As such, lymphocyte and atypical lymphocyte counts were both significantly highest in the groups that received EFA at d 1 PP. Moreover, EFA supplementation increased the mean corpuscular volume and showed a trend for induction of mean corpuscular hemoglobin compared with the CLA group during the transition period. The PP mean thrombocyte volume was higher in the EFA than in the CLA group (except for d 28) and both EFA and CLA reduced number of thrombocytes and thrombocrit at distinct time points. Hepatic mRNA abundance of markers related to oxidative status, including glutathione peroxidase (GPX-1) and catalase (CAT), was lower (P < 0.05) in EFA-treated than non-EFA-treated cows at d 28 PP. Dairy cows at the onset of lactation were characterized by induced markers of both oxidative stress and inflammation. Supplementing EFA and CLA had minor and time-dependent effects on markers of oxidative stress in plasma, erythrocytes, and liver. A comparison of EFA supplementation with CLA or CTRL showed higher immunohematological response at d 1 PP and lower hepatic antioxidant levels by d 28 PP. Supplementation with EFA+CLA had only a minor effect on oxidative markers, which were more similar to those with the EFA treatment. Altogether, despite the time-dependent differences, the current findings show only minor effects of EFA and CLA supplementation in the prevention of early lactation-induced oxidative stress.
Collapse
Affiliation(s)
- A Veshkini
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - M Gnott
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - L Vogel
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - C Kröger-Koch
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - A Tuchscherer
- Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | | | - U Bernabucci
- Department of Agronomic and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy
| | - E Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - A Starke
- Clinic for Ruminants and Swine, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - M Mielenz
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - L Bachmann
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; Faculty of Agriculture and Food Sciences, University of Applied Science Neubrandenburg, 17033 Neubrandenburg, Germany
| | - H M Hammon
- Institute of Nutritional Physiology "Oskar Kellner," Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| |
Collapse
|
50
|
Vital EF, Lam WA. Hidden behind thromboinflammation: revealing the roles of von Willebrand factor in sickle cell disease pathophysiology. Curr Opin Hematol 2023; 30:86-92. [PMID: 36853830 PMCID: PMC10065920 DOI: 10.1097/moh.0000000000000755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the pathophysiology of sickle cell disease (SCD) with a particular focus on the dysregulation of the von Willebrand factor (VWF) - ADAMTS13 axis that contributes to its pathogenesis. In discussing recent developments, we hope to encourage new and ongoing discussions surrounding therapeutic targets for SCD. RECENT FINDINGS Within the last 5 years, the role of VWF in the pathophysiology of SCD has been further elucidated and is now a target of study in ongoing clinical trials. SUMMARY The pathophysiology of SCD is multifaceted, as it involves systemwide vascular activation, altered blood rheology, and the activation of immune responses and coagulative pathways. The presence of VWF in excess in SCD, particularly in its largest multimeric form, greatly contributes to its pathogenesis. Understanding the molecular mechanisms that underly the presence of large VWF multimers in SCD will provide further insight into the pathogenesis of SCD and provide specific targets for therapy.
Collapse
Affiliation(s)
- Eudorah F. Vital
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|