1
|
Gao C, Dai Y, Spezza PA, Boasiako P, Tang A, Rasquinha G, Zhong H, Shao B, Liu Y, Shi PA, Lobo CA, An X, Guo A, Mitchell WB, Manwani D, Yazdanbakhsh K, Mendelson A. Megakaryocytes transfer mitochondria to bone marrow mesenchymal stromal cells to lower platelet activation. J Clin Invest 2025; 135:e189801. [PMID: 40014405 PMCID: PMC11996913 DOI: 10.1172/jci189801] [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: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Newly produced platelets acquire a low activation state, but whether the megakaryocyte plays a role in this outcome has not been fully uncovered. Mesenchymal stem cells (MSCs) were previously shown to promote platelet production and lower platelet activation. We found that healthy megakaryocytes transfer mitochondria to MSCs, which is mediated by connexin 43 (Cx43) gap junctions on MSCs and leads to platelets at a low energetic state with increased LYN activation, characteristic of resting platelets with increased LYN activation, characteristic of resting platelets. On the contrary, MSCs have a limited ability to transfer mitochondria to megakaryocytes. Sickle cell disease (SCD) is characterized by hemolytic anemia and results in heightened platelet activation, contributing to numerous disease complications. Platelets in SCD mice and human samples had a heightened energetic state with increased glycolysis. MSC exposure to heme in SCD led to decreased Cx43 expression and a reduced ability to uptake mitochondria from megakaryocytes. This prevented LYN activation in platelets and contributed to increased platelet activation at steady state. Altogether, our findings demonstrate an effect of hemolysis in the microenvironment leading to increased platelet activation in SCD. These findings have the potential to inspire new therapeutic targets to relieve thrombosis-related complications of SCD and other hemolytic conditions.
Collapse
Affiliation(s)
| | - Yitian Dai
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul A. Spezza
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul Boasiako
- Laboratory of Stem Cell Biology and Engineering Research
| | - Alice Tang
- Laboratory of Stem Cell Biology and Engineering Research
| | | | | | - Bojing Shao
- Laboratory of Vascular Inflammation and Thrombosis Research
| | | | | | - Cheryl A. Lobo
- Laboratory of Blood Borne Parasites, New York Blood Center, New York, New York, USA
| | | | - Anqi Guo
- Laboratory of Complement Biology
| | - William B. Mitchell
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | | | | |
Collapse
|
2
|
Vats R, Ungalara R, Dubey RK, Sundd P, Pradhan-Sundd T. Heme-Oxygenase 1 Mediated Activation of Cyp3A11 Protects Against Non-Steroidal Pain Analgesics Induced Acute Liver Damage in Sickle Cell Disease Mice. Cells 2025; 14:194. [PMID: 39936985 PMCID: PMC11817884 DOI: 10.3390/cells14030194] [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: 12/18/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Pain constitutes a significant comorbidity associated with sickle cell disease (SCD). Analgesics serve as the primary method for pain management; however, the long-term effects of these drugs on the liver of SCD patients remain not completely understood. Using real-time intravital imaging, we analyzed the effect of non-steroidal analgesics (NSA) in the liver of control and SS (SCD) mice. Remarkably, we found completely opposing effects in the liver of control and SS mice post-NSA treatment. Whereas SS mice were able to better tolerate the NSA treatment acutely compared to their littermate controls, in the long term, these mice showed delayed resolution of liver injury and exacerbated fibrosis compared to control mice. Mechanistically, we found that SS mice were protected from cytotoxicity caused by NSA at baseline due to the significant activation of hepatic Kupffer cells, which produced heme-oxygenase 1 (HO-1). HO-1 promoted the activation of the cytoprotective enzyme Cyp3A11, which inhibited hepatic damage caused by NSA. However, in the long term, depletion of hepatic Kupffer cells led to reduced expression of HO-1, which blocked the activation of Cyp3A11, resulting in fibrosis and a delay in the resolution of liver injury and inflammation. These preclinical data provide a strong proof-of-concept for HO-1 as well as Cyp3A11 as cytoprotectors against NSA-induced liver damage in the Townes model of SCD and support further development of these compounds as potential novel therapies for end-organ damage in SCD.
Collapse
Affiliation(s)
- Ravi Vats
- Versiti Blood Research Institute and Blood Center of Wisconsin, 8733 West Watertown Plank Road, Milwaukee, WI 53226, USA; (R.V.); (R.U.); (R.K.D.); (P.S.)
- Divisions of Cell Biology, Neurobiology and Anatomy and Bioengineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ramakrishna Ungalara
- Versiti Blood Research Institute and Blood Center of Wisconsin, 8733 West Watertown Plank Road, Milwaukee, WI 53226, USA; (R.V.); (R.U.); (R.K.D.); (P.S.)
- Divisions of Cell Biology, Neurobiology and Anatomy and Bioengineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rikesh K. Dubey
- Versiti Blood Research Institute and Blood Center of Wisconsin, 8733 West Watertown Plank Road, Milwaukee, WI 53226, USA; (R.V.); (R.U.); (R.K.D.); (P.S.)
- Divisions of Cell Biology, Neurobiology and Anatomy and Bioengineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Prithu Sundd
- Versiti Blood Research Institute and Blood Center of Wisconsin, 8733 West Watertown Plank Road, Milwaukee, WI 53226, USA; (R.V.); (R.U.); (R.K.D.); (P.S.)
- Divisions of Cell Biology, Neurobiology and Anatomy and Bioengineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tirthadipa Pradhan-Sundd
- Versiti Blood Research Institute and Blood Center of Wisconsin, 8733 West Watertown Plank Road, Milwaukee, WI 53226, USA; (R.V.); (R.U.); (R.K.D.); (P.S.)
- Divisions of Cell Biology, Neurobiology and Anatomy and Bioengineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
3
|
Mestekemper AN, Pirschel W, Krieg N, Paulmann MK, Daniel C, Amann K, Coldewey SM. Reduction in Renal Heme Oxygenase-1 Is Associated with an Aggravation of Kidney Injury in Shiga Toxin-Induced Murine Hemolytic-Uremic Syndrome. Toxins (Basel) 2024; 16:543. [PMID: 39728801 PMCID: PMC11679022 DOI: 10.3390/toxins16120543] [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: 10/11/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Hemolytic-uremic syndrome (HUS) is a systemic complication of an infection with Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli, primarily leading to acute kidney injury (AKI) and microangiopathic hemolytic anemia. Although free heme has been found to aggravate renal damage in hemolytic diseases, the relevance of the heme-degrading enzyme heme oxygenase-1 (HO-1, encoded by Hmox1) in HUS has not yet been investigated. We hypothesized that HO-1, also important in acute phase responses in damage and inflammation, contributes to renal pathogenesis in HUS. The effect of tamoxifen-induced Hmox1 gene deletion on renal HO-1 expression, disease progression and AKI was investigated in mice 7 days after HUS induction. Renal HO-1 levels were increased in Stx-challenged mice with tamoxifen-induced Hmox1 gene deletion (Hmox1R26Δ/Δ) and control mice (Hmox1lox/lox). This HO-1 induction was significantly lower (-43%) in Hmox1R26Δ/Δ mice compared to Hmox1lox/lox mice with HUS. Notably, the reduced renal HO-1 expression was associated with an exacerbation of kidney injury in mice with HUS as indicated by a 1.7-fold increase (p = 0.02) in plasma neutrophil gelatinase-associated lipocalin (NGAL) and a 1.3-fold increase (p = 0.06) in plasma urea, while other surrogate parameters for AKI (e.g., periodic acid Schiff staining, kidney injury molecule-1, fibrin deposition) and general disease progression (HUS score, weight loss) remained unchanged. These results indicate a potentially protective role of HO-1 in the pathogenesis of Stx-mediated AKI in HUS.
Collapse
Affiliation(s)
- Antonio N. Mestekemper
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.N.M.); (N.K.); (M.K.P.)
- Septomics Research Center, Jena University Hospital, 07745 Jena, Germany
| | - Wiebke Pirschel
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.N.M.); (N.K.); (M.K.P.)
- Septomics Research Center, Jena University Hospital, 07745 Jena, Germany
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.N.M.); (N.K.); (M.K.P.)
- Septomics Research Center, Jena University Hospital, 07745 Jena, Germany
| | - Maria K. Paulmann
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.N.M.); (N.K.); (M.K.P.)
- Septomics Research Center, Jena University Hospital, 07745 Jena, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.D.); (K.A.)
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.D.); (K.A.)
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.N.M.); (N.K.); (M.K.P.)
- Septomics Research Center, Jena University Hospital, 07745 Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
4
|
Navaneethabalakrishnan S, An X, Vinchi F. Heme- and iron-activated macrophages in sickle cell disease: an updated perspective. Curr Opin Hematol 2024; 31:275-284. [PMID: 39046855 PMCID: PMC11427154 DOI: 10.1097/moh.0000000000000836] [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] [Indexed: 07/27/2024]
Abstract
PURPOSE OF REVIEW Sickle cell disease (SCD) is a hereditary blood disorder due to a single-point mutation in the β-globin gene. The ensuing hemoglobin has the tendency to polymerize upon deoxygenation, leading to the typical sickle shape of red blood cells. While the primary pathology of sickle cell disease is a direct consequence of altered red blood cells, emerging evidence highlights the central role of macrophages in mediating hemoglobin scavenging, perpetuating oxidative stress and inflammation, and causing endothelial dysfunction and tissue remodeling. RECENT FINDINGS Recent research uncovered the impact of heme and iron overload on macrophage polarization and functions in sickle cell disease, and its implication for chronic inflammation and tissue damage in vital organs such as the liver, spleen, lungs and kidneys. By providing a thorough understanding of the dynamic interactions between macrophages and various cellular components within the sickle cell disease milieu, these studies have laid the foundation for the identification of macrophage-related cellular and molecular mechanisms potentially targetable for therapeutic purposes to attenuate sickle complications. SUMMARY This review provides a current update about recent discoveries on heme/iron-activated macrophages in SCD, shedding light on their critical role in disease pathophysiology. Ultimately, it proposes avenues for future research aimed at addressing the relevance of these cells for other sickle complications and at targeting them to mitigate disease morbidity and improve patient outcomes.
Collapse
Affiliation(s)
| | - Xiuli An
- Laboratory of Membrane Biology, Lindsley Kimball Research Institute, New York Blood Center
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley Kimball Research Institute, New York Blood Center
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
5
|
Gingell L, Hrinczenko B. Characterizing the Immature Immunophenotype of Sickle Cell Disease Monocytes. Cureus 2024; 16:e60703. [PMID: 38899253 PMCID: PMC11186669 DOI: 10.7759/cureus.60703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Sickle cell disease (SCD) is marked by episodic vaso-occlusive crisis (VOC). Recurrent VOC creates a pro-inflammatory state that induces phenotypic alterations in innate immune cells. Monocytes are of particular interest to VOC pathophysiology because they are especially malleable to inflammatory signaling. Indeed, inflammatory disease states such as chronic obstructive pulmonary disease (COPD), obesity and atherosclerosis are known to influence monocyte development and alter monocyte subpopulations. In this study, we describe SCD monocyte subsets by performing immunophenotypic flow cytometric, enzymatic, and morphologic analysis on peripheral blood. Herein, we add to the growing body of evidence suggesting aberrant monocyte populations underpin VOC pathophysiology. We found that SCD monocytes possess an immature phenotype as demonstrated by 1) decreased CD4 positivity (p < .01), 2) low α-naphthyl butyrate esterase (ANBE) expression, and 3) naïve morphologic features. We additionally found an increase in CD14+CD16-CD4- monocytes (p < .01), a subset associated with the impaired immune response of post-trauma patients. Interestingly, we also found a large proportion of CD14+CD4-HLA-DR- monocytes which, under normal circumstances, are exclusively found in neonates (p < .01). Finally, we report an increase in nonclassical monocytes (CD14dimCD16+), a subset recently shown to have a critical role in prevention and recovery from VOC.
Collapse
Affiliation(s)
- Luke Gingell
- Medical School, Michigan State University, Grand Rapids, USA
| | - Borys Hrinczenko
- Hematology/Oncology, Michigan State University, East Lansing, USA
| |
Collapse
|
6
|
Shi H, Gao L, Kirby N, Shao B, Shan X, Kudo M, Silasi R, McDaniel JM, Zhou M, McGee S, Jing W, Lupu F, Cleuren A, George JN, Xia L. Clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in sickle cell anemia mice. Blood 2024; 143:1293-1309. [PMID: 38142410 PMCID: PMC10997916 DOI: 10.1182/blood.2023021583] [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: 06/21/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Although it is caused by a single-nucleotide mutation in the β-globin gene, sickle cell anemia (SCA) is a systemic disease with complex, incompletely elucidated pathologies. The mononuclear phagocyte system plays critical roles in SCA pathophysiology. However, how heterogeneous populations of hepatic macrophages contribute to SCA remains unclear. Using a combination of single-cell RNA sequencing and spatial transcriptomics via multiplexed error-robust fluorescence in situ hybridization, we identified distinct macrophage populations with diversified origins and biological functions in SCA mouse liver. We previously found that administering the von Willebrand factor (VWF)-cleaving protease ADAMTS13 alleviated vaso-occlusive episode in mice with SCA. Here, we discovered that the ADAMTS13-cleaved VWF was cleared from the circulation by a Clec4f+Marcohigh macrophage subset in a desialylation-dependent manner in the liver. In addition, sickle erythrocytes were phagocytized predominantly by Clec4f+Marcohigh macrophages. Depletion of macrophages not only abolished the protective effect of ADAMTS13 but exacerbated vaso-occlusive episode in mice with SCA. Furthermore, promoting macrophage-mediated VWF clearance reduced vaso-occlusion in SCA mice. Our study demonstrates that hepatic macrophages are important in the pathogenesis of SCA, and efficient clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in SCA mice.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Nicole Kirby
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Mariko Kudo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - John Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Meixiang Zhou
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Wei Jing
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - James N. George
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
7
|
Yeudall S, Upchurch CM, Leitinger N. The clinical relevance of heme detoxification by the macrophage heme oxygenase system. Front Immunol 2024; 15:1379967. [PMID: 38585264 PMCID: PMC10995405 DOI: 10.3389/fimmu.2024.1379967] [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: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Heme degradation by the heme oxygenase (HMOX) family of enzymes is critical for maintaining homeostasis and limiting heme-induced tissue damage. Macrophages express HMOX1 and 2 and are critical sites of heme degradation in healthy and diseased states. Here we review the functions of the macrophage heme oxygenase system and its clinical relevance in discrete groups of pathologies where heme has been demonstrated to play a driving role. HMOX1 function in macrophages is essential for limiting oxidative tissue damage in both acute and chronic hemolytic disorders. By degrading pro-inflammatory heme and releasing anti-inflammatory molecules such as carbon monoxide, HMOX1 fine-tunes the acute inflammatory response with consequences for disorders of hyperinflammation such as sepsis. We then discuss divergent beneficial and pathological roles for HMOX1 in disorders such as atherosclerosis and metabolic syndrome, where activation of the HMOX system sits at the crossroads of chronic low-grade inflammation and oxidative stress. Finally, we highlight the emerging role for HMOX1 in regulating macrophage cell death via the iron- and oxidation-dependent form of cell death, ferroptosis. In summary, the importance of heme clearance by macrophages is an active area of investigation with relevance for therapeutic intervention in a diverse array of human diseases.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Clint M. Upchurch
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
8
|
Pfefferlé M, Vallelian F. Transcription Factor NRF2 in Shaping Myeloid Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:159-195. [PMID: 39017844 DOI: 10.1007/978-3-031-62731-6_8] [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: 07/18/2024]
Abstract
NFE2-related factor 2 (NRF2) is a master transcription factor (TF) that coordinates key cellular homeostatic processes including antioxidative responses, autophagy, proteostasis, and metabolism. The emerging evidence underscores its significant role in modulating inflammatory and immune processes. This chapter delves into the role of NRF2 in myeloid cell differentiation and function and its implication in myeloid cell-driven diseases. In macrophages, NRF2 modulates cytokine production, phagocytosis, pathogen clearance, and metabolic adaptations. In dendritic cells (DCs), it affects maturation, cytokine production, and antigen presentation capabilities, while in neutrophils, NRF2 is involved in activation, migration, cytokine production, and NETosis. The discussion extends to how NRF2's regulatory actions pertain to a wide array of diseases, such as sepsis, various infectious diseases, cancer, wound healing, atherosclerosis, hemolytic conditions, pulmonary disorders, hemorrhagic events, and autoimmune diseases. The activation of NRF2 typically reduces inflammation, thereby modifying disease outcomes. This highlights the therapeutic potential of NRF2 modulation in treating myeloid cell-driven pathologies.
Collapse
Affiliation(s)
- Marc Pfefferlé
- Department of Internal Medicine, Spital Limmattal, Schlieren, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University of Zurich and University Hospital of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Fogarty H, Ahmad A, Atiq F, Doherty D, Ward S, Karampini E, Rehill A, Leon G, Byrne C, Geoghegan R, Conroy H, Byrne M, Budde U, Schneppenheim S, Sheehan C, Ngwenya N, Baker RI, Preston RJS, Tuohy E, McMahon C, O’Donnell JS. VWF-ADAMTS13 axis dysfunction in children with sickle cell disease treated with hydroxycarbamide vs blood transfusion. Blood Adv 2023; 7:6974-6989. [PMID: 37773926 PMCID: PMC10690561 DOI: 10.1182/bloodadvances.2023010824] [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: 05/26/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Previous studies have reported elevated von Willebrand factor (VWF) levels in patients with sickle cell disease (SCD) and demonstrated a key role for the VWF-ADAMTS13 axis in the pathobiology of SCD vaso-occlusion. Although blood transfusion is the gold standard for stroke prevention in SCD, the biological mechanisms underpinning its improved efficacy compared with hydroxycarbamide are not fully understood. We hypothesized that the improved efficacy of blood transfusion might relate to differences in VWF-ADAMTS13 axis dysfunction. In total, 180 children with a confirmed diagnosis of SCD (hemoglobin SS) on hydroxycarbamide (n = 96) or blood transfusion (n = 84) were included. Despite disease-modifying treatment, plasma VWF and VWF propeptide were elevated in a significant proportion of children with SCD (33% and 47%, respectively). Crucially, all VWF parameters were significantly higher in the hydroxycarbamide compared with the blood transfusion cohort (P < .05). Additionally, increased levels of other Weibel-Palade body-stored proteins, including factor VIII (FVIII), angiopoietin-2, and osteoprotegerin were observed, indicated ongoing endothelial cell activation. Children treated with hydroxycarbamide also had higher FVIII activity and enhanced thrombin generation compared with those in the blood transfusion cohort (P < .001). Finally, hemolysis markers strongly correlated with VWF levels (P < .001) and were significantly reduced in the blood transfusion cohort (P < .001). Cumulatively, to our knowledge, our findings demonstrate for the first time that despite treatment, ongoing dysfunction of the VWF-ADAMTS13 axis is present in a significant subgroup of pediatric patients with SCD, especially those treated with hydroxycarbamide.
Collapse
Affiliation(s)
- Helen Fogarty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Azaz Ahmad
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ferdows Atiq
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Soracha Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aisling Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Byrne
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rosena Geoghegan
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Helena Conroy
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Mary Byrne
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
| | - Ulrich Budde
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Sonja Schneppenheim
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Ciara Sheehan
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Noel Ngwenya
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Ross I. Baker
- Western Australia Centre for Thrombosis and Haemostasis, Perth Blood Institute, Murdoch University, Perth, WA, Australia
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Emma Tuohy
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Corrina McMahon
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| |
Collapse
|
10
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
11
|
Liu Y, Su S, Shayo S, Bao W, Pal M, Dou K, Shi PA, Aygun B, Campbell-Lee S, Lobo CA, Mendelson A, An X, Manwani D, Zhong H, Yazdanbakhsh K. Hemolysis dictates monocyte differentiation via two distinct pathways in sickle cell disease vaso-occlusion. J Clin Invest 2023; 133:e172087. [PMID: 37490346 PMCID: PMC10503794 DOI: 10.1172/jci172087] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
Sickle cell disease (SCD) is a hereditary hemoglobinopathy characterized by painful vaso-occlusive crises (VOC) and chronic hemolysis. The mononuclear phagocyte system is pivotal to SCD pathophysiology, but the mechanisms governing monocyte/macrophage differentiation remain unknown. This study examined the influence of hemolysis on circulating monocyte trajectories in SCD. We discovered that hemolysis stimulated CSF-1 production, partly by endothelial cells via Nrf2, promoting classical monocyte (CMo) differentiation into blood patrolling monocytes (PMo) in SCD mice. However, hemolysis also upregulated CCL-2 through IFN-I, inducing CMo transmigration and differentiation into tissue monocyte-derived macrophages. Blocking CMo transmigration by anti-P selectin antibody in SCD mice increased circulating PMo, corroborating that CMo-to-tissue macrophage differentiation occurs at the expense of CMo-to-blood PMo differentiation. We observed a positive correlation between plasma CSF-1/CCL-2 ratios and blood PMo levels in patients with SCD, underscoring the clinical significance of these two opposing factors in monocyte differentiation. Combined treatment with CSF-1 and anti-P selectin antibody more effectively increased PMo numbers and reduced stasis compared with single-agent therapies in SCD mice. Altogether, these data indicate that monocyte fates are regulated by the balance between two heme pathways, Nrf2/CSF-1 and IFN-I/CCL-2, and suggest that the CSF-1/CCL-2 ratio may present a diagnostic and therapeutic target in SCD.
Collapse
Affiliation(s)
| | - Shan Su
- Laboratory of Complement Biology
| | | | | | | | - Kai Dou
- Laboratory of Immune Regulation, and
| | - Patricia A. Shi
- Clinical Research in Sickle Cell Disease, New York Blood Center, New York, New York, USA
| | - Banu Aygun
- Cohen Children’s Medical Center, New Hyde Park, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Sally Campbell-Lee
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, New York, New York, USA
| | - Hui Zhong
- Laboratory of Immune Regulation, and
| | | |
Collapse
|
12
|
Genotyping and the Future of Transfusion in Sickle Cell Disease. Hematol Oncol Clin North Am 2022; 36:1271-1284. [DOI: 10.1016/j.hoc.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
13
|
Guo Y, Zhao H, Lin Z, Ye T, Xu D, Zeng Q. Heme in Cardiovascular Diseases: A Ubiquitous Dangerous Molecule Worthy of Vigilance. Front Cell Dev Biol 2022; 9:781839. [PMID: 35127704 PMCID: PMC8807526 DOI: 10.3389/fcell.2021.781839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Heme, the protoporphyrin IX iron complex is widely present in the human body and it is involved in oxygen storage, electron transfer, and enzymatic reactions. However, free heme can be toxic as it catalyzes the production of reactive oxygen species, oxidizes lipids and proteins, and causes DNA damage, thereby inducing a pro-inflammatory environment. The generation, metabolism, and degradation of heme in the human body are regulated by precise mechanisms to ensure that heme remains non-toxic. However, in several types of cardiovascular diseases, impaired metabolism and exposure to heme may occur in pathological processes, including neovascularization, internal hemorrhage, ischemia, and reperfusion. Based on years of research, in this review, we aimed to summarize the underlying mechanisms by which heme contributes to the development of cardiovascular diseases through oxidative stress, relative pathway gene expression regulation and phenotypic changes in cells. Excess heme plays a detrimental role in atherosclerosis, heart failure, myocardial ischemia-reperfusion injury, degenerative aortic valve stenosis, cardiac iron overload. Recent researches revealed that in some cases heme involved in cardiac damage though ferroptosis. Thus, heme concentrations beyond normal levels are dangerous. Further research on the role of heme in cardiovascular diseases is needed.
Collapse
Affiliation(s)
- Yuyang Guo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hengli Zhao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Zhibin Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Taochun Ye
- Department of Cardiopulmonary Rehabilitation, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng, ; Dingli Xu,
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
- *Correspondence: Qingchun Zeng, ; Dingli Xu,
| |
Collapse
|
14
|
Buehler PW, Swindle D, Pak DI, Fini MA, Hassell K, Nuss R, Wilkerson RB, D’Alessandro A, Irwin DC. Murine models of sickle cell disease and beta-thalassemia demonstrate pulmonary hypertension with distinctive features. Pulm Circ 2021; 11:20458940211055996. [PMID: 34777785 PMCID: PMC8579334 DOI: 10.1177/20458940211055996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 01/26/2023] Open
Abstract
Sickle cell anemia and β-thalassemia intermedia are very different genetically determined hemoglobinopathies predisposing to pulmonary hypertension. The etiologies responsible for the associated development of pulmonary hypertension in both diseases are multi-factorial with extensive mechanistic contributors described. Both sickle cell anemia and β-thalassemia intermedia present with intra and extravascular hemolysis. And because sickle cell anemia and β-thalassemia intermedia share features of extravascular hemolysis, macrophage iron excess and anemia we sought to characterize the common features of the pulmonary hypertension phenotype, cardiac mechanics, and function as well as lung and right ventricular metabolism. Within the concept of iron, we have defined a unique pulmonary vascular iron accumulation in lungs of sickle cell anemia pulmonary hypertension patients at autopsy. This observation is unlike findings in idiopathic or other forms of pulmonary arterial hypertension. In this study, we hypothesized that a common pathophysiology would characterize the pulmonary hypertension phenotype in sickle cell anemia and β-thalassemia intermedia murine models. However, unlike sickle cell anemia, β-thalassemia is also a disease of dyserythropoiesis, with increased iron absorption and cellular iron extrusion. This process is mediated by high erythroferrone and low hepcidin levels as well as dysregulated iron transport due transferrin saturation, so there may be differences as well. Herein we describe common and divergent features of pulmonary hypertension in aged Berk-ss (sickle cell anemia) and Hbbth/3+ (intermediate β-thalassemia) mice and suggest translational utility as proof-of-concept models to study pulmonary hypertension therapeutics specific to genetic anemias.
Collapse
Affiliation(s)
- Paul W. Buehler
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Center for Blood Oxygen Transport, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
- Paul W. Buehler, Department of Pathology University of Maryland School of Medicine, HSF III, 8th Floor, Room 8180, Baltimore, MD 21201, USA. David C. Irwin, Department of Cardiology, University of Colorado Anschutz, Medical Campus Research Building 2, B133, Room 8121 Aurora, Colorado 80045, USA.
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - David I. Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Mehdi A. Fini
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn Hassell
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Rachelle Nuss
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Rebecca B. Wilkerson
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Angelo D’Alessandro
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - David C. Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
15
|
Moreno-Cañadas R, Luque-Martín L, Arroyo AG. Intravascular Crawling of Patrolling Monocytes: A Lèvy-Like Motility for Unique Search Functions? Front Immunol 2021; 12:730835. [PMID: 34603307 PMCID: PMC8485030 DOI: 10.3389/fimmu.2021.730835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Patrolling monocytes (PMo) are the organism’s preeminent intravascular guardians by their continuous search of damaged endothelial cells and harmful microparticles for their removal and to restore homeostasis. This surveillance is accomplished by PMo crawling on the apical side of the endothelium through regulated interactions of integrins and chemokine receptors with their endothelial ligands. We propose that the search mode governs the intravascular motility of PMo in vivo in a similar way to T cells looking for antigen in tissues. Signs of damage to the luminal side of the endothelium (local death, oxidized LDL, amyloid deposits, tumor cells, pathogens, abnormal red cells, etc.) will change the diffusive random towards a Lèvy-like crawling enhancing their recognition and clearance by PMo damage receptors as the integrin αMβ2 and CD36. This new perspective can help identify new actors to promote unique PMo intravascular actions aimed at maintaining endothelial fitness and combating harmful microparticles involved in diseases as lung metastasis, Alzheimer’s angiopathy, vaso-occlusive disorders, and sepsis.
Collapse
Affiliation(s)
- Rocío Moreno-Cañadas
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Laura Luque-Martín
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Alicia G Arroyo
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
16
|
Type I interferon is induced by hemolysis and drives antibody-mediated erythrophagocytosis in sickle cell disease. Blood 2021; 138:1162-1171. [PMID: 34166491 DOI: 10.1182/blood.2021011629] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/01/2021] [Indexed: 11/20/2022] Open
Abstract
Patients with sickle cell disease (SCD) suffer from intravascular hemolysis associated vascular injury and tissue damage. Classical monocytes (CMo), which are the most abundant of circulating monocytes, are activated in SCD, but the cause and consequences of activation remain incompletely understood. We found a positive correlation between total plasma heme levels and circulating IFN-α in patients with SCD along with upregulation of the type I Interferon (IFN-I) inducible genes in sort-purified SCD patients' CMo by transcriptome analysis. We demonstrated that hemolysis led to IFN-I expression, predominantly by mouse liver monocyte and macrophages (Mϕ), primarily through Tank kinase binding 1 (TBK1)/IκB kinase-ε (IKKε) but not TLR4. In response to hemolysis-induced IFN-I, mouse CMo migrated to the liver and differentiated into monocyte derived Mϕ, increasing their numbers by 6-fold with acute hemin treatment. Hemolysis-driven IFN-I activity also led to the induction of Fc receptor CD64 expression on monocyte and Mϕ populations, enhancing alloantibody-mediated erythrophagocytosis in SCD both in vivo in mice and in in vitro human cultures. Altogether, these data demonstrate IFN-I response to hemolysis as a novel activation pathway in monocytes and Mϕ in SCD, opening the possibility for development of IFN-I-based diagnostics and therapeutics against alloantibody-mediated erythrophagocytosis.
Collapse
|
17
|
Critical Role of Hemopexin Mediated Cytoprotection in the Pathophysiology of Sickle Cell Disease. Int J Mol Sci 2021; 22:ijms22126408. [PMID: 34203861 PMCID: PMC8232622 DOI: 10.3390/ijms22126408] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating hemopexin is the primary protein responsible for the clearance of heme; therefore, it is a systemic combatant against deleterious inflammation and oxidative stress induced by the presence of free heme. This role of hemopexin is critical in hemolytic pathophysiology. In this review, we outline the current research regarding how the dynamic activity of hemopexin is implicated in sickle cell disease, which is characterized by a pathological aggregation of red blood cells and excessive hemolysis. This pathophysiology leads to symptoms such as acute kidney injury, vaso-occlusion, ischemic stroke, pain crises, and pulmonary hypertension exacerbated by the presence of free heme and hemoglobin. This review includes in vivo studies in mouse, rat, and guinea pig models of sickle cell disease, as well as studies in human samples. In summary, the current research indicates that hemopexin is likely protective against these symptoms and that rectifying depleted hemopexin in patients with sickle cell disease could improve or prevent the symptoms. The data compiled in this review suggest that further preclinical and clinical research should be conducted to uncover pathways of hemopexin in pathological states to evaluate its potential clinical function as both a biomarker and therapy for sickle cell disease and related hemoglobinopathies.
Collapse
|
18
|
Anurogo D, Yuli Prasetyo Budi N, Thi Ngo MH, Huang YH, Pawitan JA. Cell and Gene Therapy for Anemia: Hematopoietic Stem Cells and Gene Editing. Int J Mol Sci 2021; 22:ijms22126275. [PMID: 34200975 PMCID: PMC8230702 DOI: 10.3390/ijms22126275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/23/2022] Open
Abstract
Hereditary anemia has various manifestations, such as sickle cell disease (SCD), Fanconi anemia, glucose-6-phosphate dehydrogenase deficiency (G6PDD), and thalassemia. The available management strategies for these disorders are still unsatisfactory and do not eliminate the main causes. As genetic aberrations are the main causes of all forms of hereditary anemia, the optimal approach involves repairing the defective gene, possibly through the transplantation of normal hematopoietic stem cells (HSCs) from a normal matching donor or through gene therapy approaches (either in vivo or ex vivo) to correct the patient’s HSCs. To clearly illustrate the importance of cell and gene therapy in hereditary anemia, this paper provides a review of the genetic aberration, epidemiology, clinical features, current management, and cell and gene therapy endeavors related to SCD, thalassemia, Fanconi anemia, and G6PDD. Moreover, we expound the future research direction of HSC derivation from induced pluripotent stem cells (iPSCs), strategies to edit HSCs, gene therapy risk mitigation, and their clinical perspectives. In conclusion, gene-corrected hematopoietic stem cell transplantation has promising outcomes for SCD, Fanconi anemia, and thalassemia, and it may overcome the limitation of the source of allogenic bone marrow transplantation.
Collapse
Affiliation(s)
- Dito Anurogo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Makassar, Makassar 90221, Indonesia
| | - Nova Yuli Prasetyo Budi
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Mai-Huong Thi Ngo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-H.H.); (J.A.P.); Tel.: +886-2-2736-1661 (ext. 3150) (Y.-H.H.); +62-812-9535-0097 (J.A.P.)
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Correspondence: (Y.-H.H.); (J.A.P.); Tel.: +886-2-2736-1661 (ext. 3150) (Y.-H.H.); +62-812-9535-0097 (J.A.P.)
| |
Collapse
|
19
|
Vinchi F, Sparla R, Passos ST, Sharma R, Vance SZ, Zreid HS, Juaidi H, Manwani D, Yazdanbakhsh K, Nandi V, Silva AMN, Agarvas AR, Fibach E, Belcher JD, Vercellotti GM, Ghoti H, Muckenthaler MU. Vasculo-toxic and pro-inflammatory action of unbound haemoglobin, haem and iron in transfusion-dependent patients with haemolytic anaemias. Br J Haematol 2021; 193:637-658. [PMID: 33723861 PMCID: PMC8252605 DOI: 10.1111/bjh.17361] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that free haem and iron exert vasculo‐toxic and pro‐inflammatory effects by activating endothelial and immune cells. In the present retrospective study, we compared serum samples from transfusion‐dependent patients with β‐thalassaemia major and intermedia, hereditary spherocytosis and sickle cell disease (SCD). Haemolysis, transfusions and ineffective erythropoiesis contribute to haem and iron overload in haemolytic patients. In all cohorts we observed increased systemic haem and iron levels associated with scavenger depletion and toxic ‘free’ species formation. Endothelial dysfunction, oxidative stress and inflammation markers were significantly increased compared to healthy donors. In multivariable logistic regression analysis, oxidative stress markers remained significantly associated with both haem‐ and iron‐related parameters, while soluble vascular cell adhesion molecule 1 (sVCAM‐1), soluble endothelial selectin (sE‐selectin) and tumour necrosis factor α (TNFα) showed the strongest association with haem‐related parameters and soluble intercellular adhesion molecule 1 (sICAM‐1), sVCAM‐1, interleukin 6 (IL‐6) and vascular endothelial growth factor (VEGF) with iron‐related parameters. While hereditary spherocytosis was associated with the highest IL‐6 and TNFα levels, β‐thalassaemia major showed limited inflammation compared to SCD. The sVCAM1 increase was significantly lower in patients with SCD receiving exchange compared to simple transfusions. The present results support the involvement of free haem/iron species in the pathogenesis of vascular dysfunction and sterile inflammation in haemolytic diseases, irrespective of the underlying haemolytic mechanism, and highlight the potential therapeutic benefit of iron/haem scavenging therapies in these conditions.
Collapse
Affiliation(s)
- Francesca Vinchi
- Iron Research Program, New York Blood Center, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany
| | - Richard Sparla
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Sara T Passos
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Richa Sharma
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - S Zebulon Vance
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Hala S Zreid
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Hesham Juaidi
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.,Pediatric Hematology, The Children's Hospital at Montefiore, New York, NY, USA
| | | | - Vijay Nandi
- Laboratory of Data Analytic Services, New York Blood Center, New York, NY, USA
| | - André M N Silva
- REQUIMTE-LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, University of Porto, Porto, Portugal
| | - Anand R Agarvas
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Eitan Fibach
- Department of Hematology, The Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Husam Ghoti
- European Center for Cancer and Cell Therapy (ECCT), Nicosia, Cyprus
| | - Martina U Muckenthaler
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany.,Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
20
|
A Unique Monocyte Transcriptome Discriminates Sickle Cell Disease From Other Hereditary Hemolytic Anemias and Shows the Particular Importance of Lipid and Interferon Signaling. Hemasphere 2021; 5:e531. [PMID: 33604514 PMCID: PMC7886403 DOI: 10.1097/hs9.0000000000000531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 12/17/2020] [Indexed: 01/17/2023] Open
|
21
|
Grunenwald A, Roumenina LT, Frimat M. Heme Oxygenase 1: A Defensive Mediator in Kidney Diseases. Int J Mol Sci 2021; 22:2009. [PMID: 33670516 PMCID: PMC7923026 DOI: 10.3390/ijms22042009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.
Collapse
Affiliation(s)
- Anne Grunenwald
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Lubka T. Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Marie Frimat
- U1167-RID-AGE, Institut Pasteur de Lille, Inserm, Univ. Lille, F-59000 Lille, France
- Nephrology Department, CHU Lille, Univ. Lille, F-59000 Lille, France
| |
Collapse
|
22
|
Sesti-Costa R, Borges MD, Lanaro C, de Albuquerque DM, Saad STO, Costa FF. Inflammatory Dendritic Cells Contribute to Regulate the Immune Response in Sickle Cell Disease. Front Immunol 2021; 11:617962. [PMID: 33613546 PMCID: PMC7890087 DOI: 10.3389/fimmu.2020.617962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Sickle cell disease (SCD), one of the most common hemoglobinopathies worldwide, is characterized by a chronic inflammatory component, with systemic release of inflammatory cytokines, due to hemolysis and vaso-occlusive processes. Patients with SCD demonstrate dysfunctional T and B lymphocyte responses, and they are more susceptible to infection. Although dendritic cells (DCs) are the main component responsible for activating and polarizing lymphocytic function, and are able to produce pro-inflammatory cytokines found in the serum of patients with SCD, minimal studies have thus far been devoted to these cells. In the present study, we identified the subpopulations of circulating DCs in patients with SCD, and found that the bloodstream of the patients showed higher numbers and percentages of DCs than that of healthy individuals. Among all the main DCs subsets, inflammatory DCs (CD14+ DCs) were responsible for this rise and correlated with higher reticulocyte count. The patients had more activated monocyte-derived DCs (mo-DCs), which produced MCP-1, IL-6, and IL-8 in culture. We found that a CD14+ mo-DC subset present in culture from some of the patients was the more activated subset and was mainly responsible for cytokine production, and this subset was also responsible for IL-17 production in co-culture with T lymphocytes. Finally, we suggest an involvement of heme oxygenase in the upregulation of CD14 in mo-DCs from the patients, indicating a potential mechanism for inducing inflammatory DC differentiation from circulating monocytes in the patients, which correlated with inflammatory cytokine production, T lymphocyte response skewing, and reticulocyte count.
Collapse
Affiliation(s)
- Renata Sesti-Costa
- Hematology and Hemotherapy Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Carolina Lanaro
- Hematology and Hemotherapy Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | | | | |
Collapse
|
23
|
Gbotosho OT, Kapetanaki MG, Kato GJ. The Worst Things in Life are Free: The Role of Free Heme in Sickle Cell Disease. Front Immunol 2021; 11:561917. [PMID: 33584641 PMCID: PMC7873693 DOI: 10.3389/fimmu.2020.561917] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hemolysis is a pathological feature of several diseases of diverse etiology such as hereditary anemias, malaria, and sepsis. A major complication of hemolysis involves the release of large quantities of hemoglobin into the blood circulation and the subsequent generation of harmful metabolites like labile heme. Protective mechanisms like haptoglobin-hemoglobin and hemopexin-heme binding, and heme oxygenase-1 enzymatic degradation of heme limit the toxicity of the hemolysis-related molecules. The capacity of these protective systems is exceeded in hemolytic diseases, resulting in high residual levels of hemolysis products in the circulation, which pose a great oxidative and proinflammatory risk. Sickle cell disease (SCD) features a prominent hemolytic anemia which impacts the phenotypic variability and disease severity. Not only is circulating heme a potent oxidative molecule, but it can act as an erythrocytic danger-associated molecular pattern (eDAMP) molecule which contributes to a proinflammatory state, promoting sickle complications such as vaso-occlusion and acute lung injury. Exposure to extracellular heme in SCD can also augment the expression of placental growth factor (PlGF) and interleukin-6 (IL-6), with important consequences to enthothelin-1 (ET-1) secretion and pulmonary hypertension, and potentially the development of renal and cardiac dysfunction. This review focuses on heme-induced mechanisms that are implicated in disease pathways, mainly in SCD. A special emphasis is given to heme-induced PlGF and IL-6 related mechanisms and their role in SCD disease progression.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maria G. Kapetanaki
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gregory J. Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Pal M, Bao W, Wang R, Liu Y, An X, Mitchell WB, Lobo CA, Minniti C, Shi PA, Manwani D, Yazdanbakhsh K, Zhong H. Hemolysis inhibits humoral B-cell responses and modulates alloimmunization risk in patients with sickle cell disease. Blood 2021; 137:269-280. [PMID: 33152749 PMCID: PMC7820872 DOI: 10.1182/blood.2020008511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
Red blood cell alloimmunization remains a barrier for safe and effective transfusions in sickle cell disease (SCD), but the associated risk factors remain largely unknown. Intravascular hemolysis, a hallmark of SCD, results in the release of heme with potent immunomodulatory activity, although its effect on SCD humoral response, specifically alloimmunization, remains unclear. Here, we found that cell-free heme suppresses human B-cell plasmablast and plasma cell differentiation by inhibiting the DOCK8/STAT3 signaling pathway, which is critical for B-cell activation, as well as by upregulating heme oxygenase 1 (HO-1) through its enzymatic byproducts, carbon monoxide and biliverdin. Whereas nonalloimmunized SCD B cells were inhibited by exogenous heme, B cells from the alloimmunized group were nonresponsive to heme inhibition and readily differentiated into plasma cells. Consistent with a differential B-cell response to hemolysis, we found elevated B-cell basal levels of DOCK8 and higher HO-1-mediated inhibition of activated B cells in nonalloimmunized compared with alloimmunized SCD patients. To overcome the alloimmunized B-cell heme insensitivity, we screened several heme-binding molecules and identified quinine as a potent inhibitor of B-cell activity, reversing the resistance to heme suppression in alloimmunized patients. B-cell inhibition by quinine occurred only in the presence of heme and through HO-1 induction. Altogether, these data suggest that hemolysis can dampen the humoral B-cell response and that B-cell heme responsiveness maybe a determinant of alloimmunization risk in SCD. By restoring B-cell heme sensitivity, quinine may have therapeutic potential to prevent and inhibit alloimmunization in SCD patients.
Collapse
Affiliation(s)
| | | | | | | | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - William B Mitchell
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, NY
| | - Cheryl A Lobo
- Laboratory of Blood-Borne Parasites, New York Blood Center, New York, NY
| | - Caterina Minniti
- Department of Medicine, Division of Hematology, Montefiore Health Center, Albert Einstein College of Medicine, Bronx, NY; and
| | - Patricia A Shi
- Sickle Cell Clinical Research Program, New York Blood Center, New York, NY
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, NY
| | | | - Hui Zhong
- Laboratory of Immune Regulation, and
| |
Collapse
|
25
|
Nath M, Agarwal A. New insights into the role of heme oxygenase-1 in acute kidney injury. Kidney Res Clin Pract 2020; 39:387-401. [PMID: 33184238 PMCID: PMC7770992 DOI: 10.23876/j.krcp.20.091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI) is attended by injury-related biomarkers appearing in the urine and serum, decreased urine output, and impaired glomerular filtration rate. AKI causes increased morbidity and mortality and can progress to chronic kidney disease and end-stage kidney failure. AKI is without specific therapies and is managed by supported care. Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. HO-1 induction protects against assorted renal insults as demonstrated by in vitro and preclinical models. This review summarizes the advances in understanding of the protection conferred by HO-1 in AKI, how HO-1 can be induced including via its transcription factor Nrf2, and HO-1 induction as a therapeutic strategy.
Collapse
Affiliation(s)
- Meryl Nath
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Veterans Affairs, Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
26
|
Allali S, Maciel TT, Hermine O, de Montalembert M. Innate immune cells, major protagonists of sickle cell disease pathophysiology. Haematologica 2020; 105:273-283. [PMID: 31919091 PMCID: PMC7012475 DOI: 10.3324/haematol.2019.229989] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/26/2019] [Indexed: 12/30/2022] Open
Abstract
Sickle cell disease (SCD), considered the most common monogenic disease worldwide, is a severe hemoglobin disorder. Although the genetic and molecular bases have long been characterized, the pathophysiology remains incompletely elucidated and therapeutic options are limited. It has been increasingly suggested that innate immune cells, including monocytes, neutrophils, invariant natural killer T cells, platelets and mast cells, have a role in promoting inflammation, adhesion and pain in SCD. Here we provide a thorough review of the involvement of these novel, major protagonists in SCD pathophysiology, highlighting recent evidence for innovative therapeutic perspectives.
Collapse
Affiliation(s)
- Slimane Allali
- Department of General Pediatrics and Pediatric Infectious Diseases, Reference Center for Sickle Cell Disease, Necker Hospital for Sick Children, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris Descartes University, Paris .,Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, Paris Descartes - Sorbonne Paris Cite University, Imagine Institute, Inserm U1163, Paris.,Laboratory of Excellence GR-Ex, Paris
| | - Thiago Trovati Maciel
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, Paris Descartes - Sorbonne Paris Cite University, Imagine Institute, Inserm U1163, Paris.,Laboratory of Excellence GR-Ex, Paris
| | - Olivier Hermine
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, Paris Descartes - Sorbonne Paris Cite University, Imagine Institute, Inserm U1163, Paris.,Laboratory of Excellence GR-Ex, Paris.,Department of Hematology, Necker Hospital for Sick Children, AP-HP, Paris Descartes University, Paris, France
| | - Mariane de Montalembert
- Department of General Pediatrics and Pediatric Infectious Diseases, Reference Center for Sickle Cell Disease, Necker Hospital for Sick Children, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris Descartes University, Paris .,Laboratory of Excellence GR-Ex, Paris
| |
Collapse
|
27
|
Guarda CC, Silveira-Mattos PSM, Yahouédéhou SCMA, Santiago RP, Aleluia MM, Figueiredo CVB, Fiuza LM, Carvalho SP, Oliveira RM, Nascimento VML, Luz NF, Borges VM, Andrade BB, Gonçalves MS. Hydroxyurea alters circulating monocyte subsets and dampens its inflammatory potential in sickle cell anemia patients. Sci Rep 2019; 9:14829. [PMID: 31616024 PMCID: PMC6794261 DOI: 10.1038/s41598-019-51339-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/17/2019] [Indexed: 01/27/2023] Open
Abstract
Sickle cell anemia (SCA) is a hemolytic disease in which vaso-occlusion is an important pathophysiological mechanism. The treatment is based on hydroxyurea (HU), which decreases leukocyte counts and increases fetal hemoglobin synthesis. Different cell types are thought to contribute to vaso-occlusion. Nevertheless, the role of monocytes subsets remains unclear. We investigated frequencies of monocytes subsets in blood and their response to HU therapy, testing their ability to express pro-inflammatory molecules and tissue factor (TF). We identified major changes in monocyte subsets, with classical monocytes (CD14++CD16−) appearing highly frequent in who were not taking HU, whereas those with patrolling phenotype (CD14dimCD16+) were enriched in individuals undergoing therapy. Additionally, HU decreased the production of TNF-α, IL1-β, IL-6, IL-8 as well as TF by the LPS-activated monocytes. Likewise, frequency of TF-expressing monocytes is increased in patients with previous vaso-occlusion. Moreover, activated monocytes expressing TF produced several pro-inflammatory cytokines simultaneously. Such polyfunctional capacity was dramatically dampened by HU therapy. The frequency of classical monocytes subset was positively correlated with percentage cytokine producing cells upon LPS stimulation. These findings suggest that classical monocytes are the subset responsible for multiple pro-inflammatory cytokine production and possibly drive inflammation and vaso-occlusion in SCA which is damped by HU.
Collapse
Affiliation(s)
- Caroline C Guarda
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Paulo S M Silveira-Mattos
- Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil.,Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Bahia, Brazil
| | - Sètondji C M A Yahouédéhou
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Rayra P Santiago
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Milena M Aleluia
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil
| | - Camylla V B Figueiredo
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Luciana M Fiuza
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Suellen P Carvalho
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Rodrigo M Oliveira
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil.,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Valma M L Nascimento
- Fundação de Hematologia e Hemoterapia do Estado da Bahia (HEMOBA) Salvador, Bahia, Brazil
| | - Nívea F Luz
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil
| | - Valéria M Borges
- Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil
| | - Bruno B Andrade
- Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil. .,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil. .,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil. .,Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Bahia, Brazil. .,Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, Brazil. .,Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil.
| | - Marilda S Gonçalves
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, FIOCRUZ-BA, Salvador, Bahia, Brazil. .,Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.
| |
Collapse
|
28
|
Balbuena-Merle R, Curtis SA, Devine L, Gibb DR, Karafin MS, Luckey CJ, Tormey CA, Siddon AJ, Roberts JD, Hendrickson JE. Red blood cell alloimmunization is associated with lower expression of FcγR1 on monocyte subsets in patients with sickle cell disease. Transfusion 2019; 59:3219-3227. [PMID: 31355970 PMCID: PMC7075520 DOI: 10.1111/trf.15463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/25/2019] [Accepted: 07/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Despite the clinical significance of red blood cell (RBC) alloantibodies, there are currently no laboratory tests available to predict which patients may be at risk of antibody formation after transfusion exposure. Given their phagocytic and inflammatory functions, we hypothesized that differences in circulating monocytes may play a role in alloimmunization. STUDY DESIGN AND METHODS Forty-two adults with sickle cell disease (SCD) were recruited, with data extracted from the electronic medical record and peripheral blood analyzed by flow cytometry for total monocytes, monocyte subsets (CD14 high/CD16 low+ classical monocytes, CD14 high/CD16 high+ intermediate monocytes, and CD14 intermediate/CD16 high+ non-classical/inflammatory monocytes), and FcγR1 (CD64) expression. Thirteen "non-responder" patients (non-alloimmunized patients with documented RBC transfusion at the study institution) were compared to 20 alloimmunized "responder" patients, who had a total of 44 RBC alloantibodies identified. RESULTS There were no significant differences in the percentages of total monocytes, monocyte subsets, or measured cytokines between non-responders and responders. However, non-responders had higher CD64 expression on classical monocytes (MFI mean 3424 ± standard deviation 1141) compared to responders (MFI mean 2285 ± 1501), p = 0.029, and on intermediate monocytes (MFI mean 3720 ± 1191) compared to responders (MFI mean 2497 ± 1640), p = 0.033. CONCLUSIONS Monocytes and the inflammatory milieu increasingly are being appreciated to play a role in some complications of SCD. The differences in FcγR1 expression on monocyte subsets noted between responders and non-responders, which cannot be directly explained by the serum cytokines evaluated, warrant further investigation.
Collapse
Affiliation(s)
| | | | - Lesley Devine
- Department of Laboratory Medicine, Yale University, New Haven
| | - David R. Gibb
- Department of Laboratory Medicine, Yale University, New Haven
| | - Matthew S. Karafin
- Medical Sciences Institute, Blood Center of Wisconsin, part of Versiti, Milwaukee, WI
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Christopher A. Tormey
- Department of Laboratory Medicine, Yale University, New Haven
- VA Connecticut, Pathology and Laboratory Medicine Service, West Haven, Connecticut
| | - Alexa J. Siddon
- Department of Laboratory Medicine, Yale University, New Haven
- VA Connecticut, Pathology and Laboratory Medicine Service, West Haven, Connecticut
- Department of Pathology, Yale University, New Haven
| | | | - Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University, New Haven
- Department of Pediatrics, Yale University, New Haven
| |
Collapse
|
29
|
Circulating Heme Oxygenase-1 and Complement Activation in Transplant-Associated Thrombotic Microangiopathy. Biol Blood Marrow Transplant 2019; 25:1486-1491. [DOI: 10.1016/j.bbmt.2019.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 03/01/2019] [Indexed: 01/08/2023]
|
30
|
Adebiyi MG, Zhao Z, Ye Y, Manalo J, Hong Y, Lee CC, Xian W, McKeon F, Culp-Hill R, D' Alessandro A, Kellems RE, Yoo SH, Han L, Xia Y. Circadian period 2: a missing beneficial factor in sickle cell disease by lowering pulmonary inflammation, iron overload, and mortality. FASEB J 2019; 33:10528-10537. [PMID: 31260634 DOI: 10.1096/fj.201900246rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian clock is important for cellular and organ function. However, its function in sickle cell disease (SCD), a life-threatening hemolytic disorder, remains unknown. Here, we performed an unbiased microarray screen, which revealed significantly altered expression of circadian rhythmic genes, inflammatory response genes, and iron metabolic genes in SCD Berkeley transgenic mouse lungs compared with controls. Given the vital role of period 2 (Per2) in the core clock and the unrecognized role of Per2 in SCD, we transplanted the bone marrow (BM) of SCD mice to Per2Luciferase mice, which revealed that Per2 expression was up-regulated in SCD mouse lung. Next, we transplanted the BM of SCD mice to period 1 (Per1)/Per2 double deficient [Per1/Per2 double knockout (dKO)] and wild-type mice, respectively. We discovered that Per1/Per2 dKO mice transplanted with SCD BM (SCD → Per1/Per2 dKO) displayed severe irradiation sensitivity and were more susceptible to an early death. Although we observed an increase of peripheral inflammatory cells, we did not detect differences in erythrocyte sickling. However, there was further lung damage due to elevated pulmonary congestion, inflammatory cell infiltration, iron overload, and secretion of IL-6 in lavage fluid. Overall, we demonstrate that Per1/Per2 is beneficial to counteract elevated systemic inflammation, lung tissue inflammation, and iron overload in SCD.-Adebiyi, M. G., Zhao, Z., Ye, Y., Manalo, J., Hong, Y., Lee, C. C., Xian, W., McKeon, F., Culp-Hill, R., D' Alessandro, A., Kellems, R. E., Yoo, S.-H., Han, L., Xia, Y. Circadian period 2: a missing beneficial factor in sickle cell disease by lowering pulmonary inflammation, iron overload, and mortality.
Collapse
Affiliation(s)
- Morayo G Adebiyi
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Zhaoyang Zhao
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Jeanne Manalo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Yue Hong
- Department of Biology and Biochemistry, The University of Houston, Houston, Texas, USA
| | - Cheng Chi Lee
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Wa Xian
- The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Frank McKeon
- Department of Biology and Biochemistry, The University of Houston, Houston, Texas, USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D' Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA.,The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA.,The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
31
|
Patrolling monocytes scavenge endothelial-adherent sickle RBCs: a novel mechanism of inhibition of vaso-occlusion in SCD. Blood 2019; 134:579-590. [PMID: 31076443 DOI: 10.1182/blood.2019000172] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/02/2019] [Indexed: 12/23/2022] Open
Abstract
Painful vaso-occlusive crisis (VOC) is the most common complication of sickle cell disease (SCD). Increasing evidence suggests that vaso-occlusion is initiated by increased adherence of sickle red blood cells (RBCs) to the vascular endothelium. Thus, the mechanisms that remove endothelial-attached sickle RBCs from the microvasculature are expected to be critical for optimal blood flow and prevention of VOC in SCD. We hypothesized that patrolling monocytes (PMos), which protect against vascular damage by scavenging cellular debris, could remove endothelial-adherent sickle RBCs and ameliorate VOC in SCD. We detected RBC (GPA+)-engulfed material in circulating PMos of patients with SCD, and their frequency was further increased during acute crisis. RBC uptake by PMos was specific to endothelial-attached sickle, but not control, RBCs and occurred mostly through ICAM-1, CD11a, and CD18. Heme oxygenase 1 induction, by counteracting the cytotoxic effects of engulfed RBC breakdown products, increased PMo viability. In addition, transfusions, by lowering sickle RBC uptake, improved PMo survival. Selective depletion of PMos in Townes sickle mice exacerbated vascular stasis and tissue damage, whereas treatment with muramyl dipeptide (NOD2 ligand), which increases PMo mass, reduced stasis and SCD associated organ damage. Altogether, these data demonstrate a novel mechanism for removal of endothelial attached sickle RBCs mediated by PMos that can protect against VOC pathogenesis, further supporting PMos as a promising therapeutic target in SCD VOC.
Collapse
|
32
|
Liu Y, Zhong H, Vinchi F, Mendelson A, Yazdanbakhsh K. Patrolling monocytes in sickle cell hemolytic conditions. Transfus Clin Biol 2019; 26:128-129. [PMID: 30898432 PMCID: PMC6488014 DOI: 10.1016/j.tracli.2019.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Patients with sickle cell disease (SCD) suffer from intravascular hemolysis associated with vascular injury and dysfunction. Painful vaso-occlusive crisis (VOC) involving increased attachment of sickle erythrocytes and activated leukocytes to damaged vascular endothelium is a hallmark of SCD. Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme with anti-cytotoxic and anti-inflammatory properties. Recent data show that patients with SCD have a novel subset of patrolling monocytes expressing very high levels of HO-1 (HO-1hi) which are decreased in numbers in patients who had a recent VOC episode. This patrolling monocyte subset was responsible for protection of endothelium against sickle RBC stasis in an experimental model. This raises the possibility that patrolling monocytes may also offer protection against vascular endothelium damage in hyperhemolytic conditions in SCD.
Collapse
Affiliation(s)
- Y Liu
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67(th) Street, NY10065 New York, United States.
| | - H Zhong
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67(th) Street, NY10065 New York, United States
| | - F Vinchi
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67(th) Street, NY10065 New York, United States
| | - A Mendelson
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67(th) Street, NY10065 New York, United States
| | - K Yazdanbakhsh
- Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67(th) Street, NY10065 New York, United States
| |
Collapse
|
33
|
A novel nonsense mutation in ADAMTS17 caused autosomal recessive inheritance Weill–Marchesani syndrome from a Chinese family. J Hum Genet 2019; 64:681-687. [DOI: 10.1038/s10038-019-0608-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/12/2019] [Indexed: 02/04/2023]
|
34
|
Altered Peripheral Blood Leucocyte Phenotype and Responses in Healthy Individuals with Homozygous Deletion of FHR1 and FHR3 Genes. J Clin Immunol 2019; 39:336-345. [DOI: 10.1007/s10875-019-00619-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/26/2019] [Indexed: 01/10/2023]
|
35
|
Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:263-292. [PMID: 30332562 DOI: 10.1146/annurev-pathmechdis-012418-012838] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the discovery of sickle cell disease (SCD) in 1910, enormous strides have been made in the elucidation of the pathogenesis of its protean complications, which has inspired recent advances in targeted molecular therapies. In SCD, a single amino acid substitution in the β-globin chain leads to polymerization of mutant hemoglobin S, impairing erythrocyte rheology and survival. Clinically, erythrocyte abnormalities in SCD manifest in hemolytic anemia and cycles of microvascular vaso-occlusion leading to end-organ ischemia-reperfusion injury and infarction. Vaso-occlusive events and intravascular hemolysis promote inflammation and redox instability that lead to progressive small- and large-vessel vasculopathy. Based on current evidence, the pathobiology of SCD is considered to be a vicious cycle of four major processes, all the subject of active study and novel therapeutic targeting: ( a) hemoglobin S polymerization, ( b) impaired biorheology and increased adhesion-mediated vaso-occlusion, ( c) hemolysis-mediated endothelial dysfunction, and ( d) concerted activation of sterile inflammation (Toll-like receptor 4- and inflammasome-dependent innate immune pathways). These molecular, cellular, and biophysical processes synergize to promote acute and chronic pain and end-organ injury and failure in SCD. This review provides an exhaustive overview of the current understanding of the molecular pathophysiology of SCD, how this pathophysiology contributes to complications of the central nervous and cardiopulmonary systems, and how this knowledge is being harnessed to develop current and potential therapies.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
36
|
How I safely transfuse patients with sickle-cell disease and manage delayed hemolytic transfusion reactions. Blood 2018; 131:2773-2781. [PMID: 29724898 DOI: 10.1182/blood-2018-02-785964] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/01/2018] [Indexed: 12/15/2022] Open
Abstract
Transfusions can be a life-saving treatment of patients with sickle-cell disease (SCD). However, availability of matched units can be limiting because of distinctive blood group polymorphisms in patients of African descent. Development of antibodies against the transfused red blood cells (RBCs), resulting in delayed hemolytic transfusion reactions (DHTRs), can be life-threatening and pose unique challenges for this population with regard to treatment strategies and transfusion management protocols. In cases where the transfused cells and the patient's own RBCs are destroyed, diagnosis of DHTR can be difficult because symptoms may mimic vaso-occlusive crisis, and frequently, antibodies are undetectable. Guidelines are needed for early diagnosis of DHTR because treatment may need to include temporarily withholding any new transfusions to avoid further hemolysis. Also needed are case-control studies to optimally tailor treatments based on the severity of DHTR and develop preventive transfusion strategies for patients at DHTR risk. Here, we will review gaps in knowledge and describe through case studies our recommended approach to prevent alloimmunization and to diagnose and treat symptomatic DHTRs for which complementary mechanistic studies to understand their pathogenesis are sorely needed.
Collapse
|
37
|
|