1
|
Aumann MA, Richerson W, Song AK, Martin D, Davis LT, Davis SM, Milner LL, Kassim AA, DeBaun MR, Jordan LC, Donahue MJ. Cerebral Hemodynamic Responses to Disease-Modifying and Curative Sickle Cell Disease Therapies. Neurology 2025; 104:e210191. [PMID: 39705613 DOI: 10.1212/wnl.0000000000210191] [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: 07/29/2024] [Accepted: 10/23/2024] [Indexed: 12/22/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Sickle cell disease (SCD) is a hemoglobinopathy resulting in hemoglobin-S production, hemolytic anemia, and elevated stroke risk. Treatments include oral hydroxyurea, blood transfusions, and hematopoietic stem cell transplantation (HSCT). Our objective was to evaluate the neurologic relevance of these therapies by characterizing how treatment-induced changes in hemoglobin (Hb) affect brain health biomarkers. METHODS In this interventional study, adults with and without SCD underwent a 3T-MRI at Vanderbilt University Medical Center at 2 time points before and after clinically indicated transfusion or HSCT or at 2 time points without the introduction of a new Hb-altering therapy (adult controls and patients with SCD on hydroxyurea). Cerebral blood flow (CBF; mL/100 g/min) and cerebral venous blood relaxation rate (s-1; a marker of Hb and blood oxygen content) responses were assessed to understand how these markers of brain health vary with Hb modulation. CBF was assessed with arterial spin labeling MRI, and blood relaxation rate was assessed using T2 relaxation under spin tagging MRI. Measures were pairwise compared within each cohort using a 2-tailed Wilcoxon signed-rank test, and regression was applied to evaluate the parameter and Hb change relationships. The significance criterion was 2-sided p < 0.05. RESULTS Adults with (n = 43; age 28.7 ± 7.7 years; 42% male) and without (n = 13; age 33.5 ± 12.2 years; 46% male) SCD were evaluated. In adults receiving hydroxyurea (n = 10), neither Hb, CBF, nor venous relaxation rate changed between time 1 (Hb = 8.6 ± 1.2 g/dL) and time 2 (Hb = 9.0 ± 1.8 g/dL) (all p > 0.05). In transfusion patients (n = 19), Hb increased from 8.2 ± 1.4 g/dL to 9.3 ± 1.3 g/dL before vs after transfusion (p < 0.001), paralleling a CBF decrease of 14.2 mL/100 g/min (p < 0.001) toward control levels. The venous relaxation rate did not change after transfusion (p = 0.71). In HSCT patients (n = 14), Hb increased from 8.9 ± 1.9 g/dL to 12.9 ± 2.7 g/dL (p < 0.001) before vs after transplant, paralleling CBF decreases from 68.16 ± 20.24 to 47.43 ± 12.59 mL/100 g/min (p < 0.001) and increase in venous relaxation rate (p = 0.004). Across the Hb spectrum, a CBF decrease of 5.02 mL/100 g/min per g/dL increase in Hb was observed. DISCUSSION Findings demonstrate improvement in cerebral hemodynamics after transfusion and transplant therapies compared with hydroxyurea therapy; quantitative relationships should provide a framework for using these measures as trial end points to assess how new SCD therapies affect brain health.
Collapse
Affiliation(s)
- Megan A Aumann
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Wesley Richerson
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Alexander K Song
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Dann Martin
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - L Taylor Davis
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Samantha M Davis
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Lauren L Milner
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Adetola A Kassim
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Michael R DeBaun
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Lori C Jordan
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| | - Manus J Donahue
- From the Department of Neurology (M.A.A., W.R., A.K.S., M.J.D.), Department of Radiology and Radiological Sciences (D.M., L.T.D., L.C.J.), Division of Pediatric Neurology, Department of Pediatrics (S.M.D., L.L.M., L.C.J.), Division of Hematology and Oncology, Department of Medicine (A.A.K., M.R.D.), and Department of Psychiatry and Behavioral Sciences (M.J.D.), Vanderbilt University Medical Center, Nashville; Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease (A.A.K., M.R.D.), Nashville; and Department of Electrical and Computer Engineering (M.J.D.), Vanderbilt University, Nashville, TN
| |
Collapse
|
2
|
Creary S, Chung MG, Villella AD, Lo WD. Stroke Prevention and Treatment for Youth with Sickle Cell Anemia: Current Practice and Challenges and Promises for the Future. Curr Neurol Neurosci Rep 2024; 24:537-546. [PMID: 39304580 PMCID: PMC11455693 DOI: 10.1007/s11910-024-01372-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE OF REVIEW Sickle cell anemia (SCA) is an autosomal recessive inherited hemoglobinopathy that results in a high risk of stroke. SCA primarily affects an underserved minority population of children who are frequently not receiving effective, multi-disciplinary, preventative care. This article reviews primary and secondary stroke prevention and treatment for children with SCA for the general adult and pediatric neurologist, who may play an important role in providing critical neurologic evaluation and care to these children. RECENT FINDINGS Primary stroke prevention is efficacious at reducing ischemic stroke risk, but it is not consistently implemented into clinical practice in the United States, resulting in these children remaining at high risk. Acute symptomatic stroke management requires neurology involvement and emergent transfusion to limit ischemia. Furthermore, while chronic transfusion therapy is a proven secondary preventative modality for those with prior symptomatic or silent cerebral infarcts, it carries significant burden. Newer therapies (e.g., stem cell therapies and voxelotor) deserve further study as they may hold promise in reducing stroke risk and treatment burden. Effective primary and secondary stroke prevention and treatment remain a challenge. Informing and engaging neurology providers to recognize and provide critical neurologic evaluation and treatment has potential to close care gaps.
Collapse
Affiliation(s)
- Susan Creary
- Division of Hematology/Oncology/BMT, Dept of Pediatrics, The Ohio State University and Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Melissa G Chung
- Division of Neurology, Dept of Pediatrics, The Ohio State University and Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
- Division of Critical Care, Dept of Pediatrics, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Anthony D Villella
- Division of Hematology/Oncology/BMT, Dept of Pediatrics, The Ohio State University and Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Warren D Lo
- Division of Neurology, Dept of Pediatrics, The Ohio State University and Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| |
Collapse
|
3
|
Dhodapkar MM, Halperin SJ, Saifi C, Whang P, Grauer JN, Varthi A. Ninety day adverse events following single level posterior lumbar interbody fusion in patients with sickle cell disease. Spine J 2024; 24:807-811. [PMID: 38081460 DOI: 10.1016/j.spinee.2023.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/21/2023] [Accepted: 11/27/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND CONTEXT Sickle cell disease (SCD) is an inherited disorder of red blood cells caused by mutation in the hemoglobin beta chain and is the most common inherited genetic disorder in the United States. Postoperative outcomes following single-level posterior lumbar interbody fusion (PLIF) in patients with SCD are not well characterized. PURPOSE To examine 90-day postoperative outcomes among a matched cohort of patients with and without SCD who underwent single level PLIF utilizing a national administrative database. STUDY DESIGN/SETTING Retrospective database study. PATIENT SAMPLE Adult patients without and with SCD who underwent single level PLIF from 2010 to 2021 Q1 were identified and matched 10:1 based on age, sex and ECI. Exclusion criteria included: age <18 years old, recent history of infection, neoplasm, or trauma, as well as not being active in the database for 90 days following their procedure. OUTCOME MEASURES Ninety-day postoperative adverse events and emergency department (ED) visits. METHODS The matched cohorts were defined, and 90-day adverse event and ED visit rates were compared with univariable analyses and multivariable logistic regression, controlling for age, sex, and Elixhauser comorbidity index (ECI). RESULTS Overall, 191,765 PLIF patients were identified, of which SCD was noted for 76 (0.04%). On multivariable analysis of the matched populations, patients with SCD were at increased odds ratio (OR) of the following (in decreasing OR order): transfusion (OR 17.69), pneumonia (OR 6.30), sepsis (OR 4.86), aggregated minor adverse events (OR 4.65), aggregated all adverse events (OR 3.87), ED visits (OR 3.53), and aggregated severe adverse events (OR 2.80) (p<.05 for all). CONCLUSIONS The current study examined a relatively small, but largest to date, cohort of SCD patients undergoing PLIF. Patients with this condition were at greater odds of several perioperative adverse events, and these findings may be helpful for patient counselling and surgical planning.
Collapse
Affiliation(s)
- Meera M Dhodapkar
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 47 College St, New Haven, CT, USA
| | - Scott J Halperin
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 47 College St, New Haven, CT, USA
| | - Comron Saifi
- Department of Orthopaedic Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Peter Whang
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 47 College St, New Haven, CT, USA
| | - Jonathan N Grauer
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 47 College St, New Haven, CT, USA
| | - Arya Varthi
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, 47 College St, New Haven, CT, USA.
| |
Collapse
|
4
|
Rarick KR, Li K, Teng RJ, Jing X, Martin DP, Xu H, Jones DW, Hogg N, Hillery CA, Garcia G, Day BW, Naylor S, Pritchard KA. Sterile inflammation induces vasculopathy and chronic lung injury in murine sickle cell disease. Free Radic Biol Med 2024; 215:112-126. [PMID: 38336101 PMCID: PMC11290318 DOI: 10.1016/j.freeradbiomed.2024.01.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Murine sickle cell disease (SCD) results in damage to multiple organs, likely mediated first by vasculopathy. While the mechanisms inducing vascular damage remain to be determined, nitric oxide bioavailability and sterile inflammation are both considered to play major roles in vasculopathy. Here, we investigate the effects of high mobility group box-1 (HMGB1), a pro-inflammatory damage-associated molecular pattern (DAMP) molecule on endothelial-dependent vasodilation and lung morphometrics, a structural index of damage in sickle (SS) mice. SS mice were treated with either phosphate-buffered saline (PBS), hE-HMGB1-BP, an hE dual-domain peptide that binds and removes HMGB1 from the circulation via the liver, 1-[4-(aminocarbonyl)-2-methylphenyl]-5-[4-(1H-imidazol-1-yl)phenyl]-1H-pyrrole-2-propanoic acid (N6022) or N-acetyl-lysyltyrosylcysteine amide (KYC) for three weeks. Human umbilical vein endothelial cells (HUVEC) were treated with recombinant HMGB1 (r-HMGB1), which increases S-nitrosoglutathione reductase (GSNOR) expression by ∼80%, demonstrating a direct effect of HMGB1 to increase GSNOR. Treatment of SS mice with hE-HMGB1-BP reduced plasma HMGB1 in SS mice to control levels and reduced GSNOR expression in facialis arteries isolated from SS mice by ∼20%. These changes were associated with improved endothelial-dependent vasodilation. Treatment of SS mice with N6022 also improved vasodilation in SS mice suggesting that targeting GSNOR also improves vasodilation. SCD decreased protein nitrosothiols (SNOs) and radial alveolar counts (RAC) and increased GSNOR expression and mean linear intercepts (MLI) in lungs from SS mice. The marked changes in pulmonary morphometrics and GSNOR expression throughout the lung parenchyma in SS mice were improved by treating with either hE-HMGB1-BP or KYC. These data demonstrate that murine SCD induces vasculopathy and chronic lung disease by an HMGB1- and GSNOR-dependent mechanism and suggest that HMGB1 and GSNOR might be effective therapeutic targets for reducing vasculopathy and chronic lung disease in humans with SCD.
Collapse
Affiliation(s)
- Kevin R Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA
| | - Keguo Li
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ru-Jeng Teng
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA
| | - Xigang Jing
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Dustin P Martin
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hao Xu
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Deron W Jones
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Cheryl A Hillery
- Department of Pediatrics, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Department of Pediatrics, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Guilherme Garcia
- Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | | | - Kirkwood A Pritchard
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; ReNeuroGen LLC, Milwaukee, WI, 53122, USA; Childrens' Research Institute, Children's Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
5
|
Sinkey RG, Ogunsile FJ, Kanter J, Bean C, Greenberg M. Society for Maternal-Fetal Medicine Consult Series #68: Sickle cell disease in pregnancy. Am J Obstet Gynecol 2024; 230:B17-B40. [PMID: 37866731 PMCID: PMC10961101 DOI: 10.1016/j.ajog.2023.10.031] [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] [Indexed: 10/24/2023]
Abstract
Pregnant individuals with sickle cell disease have an increased risk of maternal and perinatal morbidity and mortality. However, prepregnancy counseling and multidisciplinary care can lead to favorable maternal and neonatal outcomes. In this consult series, we summarize what is known about sickle cell disease and provide guidance for sickle cell disease management during pregnancy. The following are Society for Maternal-Fetal Medicine recommendations.
Collapse
|
6
|
Obeagu EI, Ubosi NI, Obeagu GU, Egba SI, Bluth MH. Understanding apoptosis in sickle cell anemia patients: Mechanisms and implications. Medicine (Baltimore) 2024; 103:e36898. [PMID: 38215146 PMCID: PMC10783340 DOI: 10.1097/md.0000000000036898] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024] Open
Abstract
Sickle cell anemia (SCA) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin, leading to the formation of sickle-shaped red blood cells. While much research has focused on the molecular and cellular mechanisms underlying the pathophysiology of SCA, recent attention has turned to the role of apoptosis, or programmed cell death, in the disease progression. This review aims to elucidate the intricate mechanisms of apoptosis in SCA patients and explore its implications in disease severity, complications, and potential therapeutic interventions. Different research search engines such as PubMed central, Scopus, Web of Science, Google Scholar, ResearchGate, Academia Edu, etc were utilized in writing this paper. Apoptosis, a highly regulated cellular process, plays a crucial role in maintaining homeostasis by eliminating damaged or dysfunctional cells. In SCA, the imbalance between pro-apoptotic and anti-apoptotic signals contributes to increased erythrocyte apoptosis, exacerbating anemia and vaso-occlusive crises. Various factors, including oxidative stress, inflammation, and altered cell signaling pathways, converge to modulate the apoptotic response in SCA. Furthermore, the interaction between apoptotic cells and the vascular endothelium contributes to endothelial dysfunction, promoting the pathogenesis of vasculopathy and organ damage seen in SCA patients. In conclusion, unraveling the complexities of apoptosis in SCA provides valuable insights into the disease pathophysiology and offers novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
| | - Nwanganga Ihuoma Ubosi
- Department of Public Health Sciences, Faculty of Health Sciences, National Open University of Nigeria, Headquarters, Jabi, Abuja, Nigeria
| | | | - Simeon Ikechukwu Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | - Martin H. Bluth
- Department of Pathology, Division of Blood Transfusion Medicine, Maimonides Medical Center, Brooklyn, NY, USA
| |
Collapse
|
7
|
Hakami F, Alhazmi E, Busayli WM, Althurwi S, Darraj AM, Alamir MA, Hakami A, Othman RA, Moafa AI, Mahasi HA, Madkhali MA. Overview of the Association Between the Pathophysiology, Types, and Management of Sickle Cell Disease and Stroke. Cureus 2023; 15:e50577. [PMID: 38107212 PMCID: PMC10723021 DOI: 10.7759/cureus.50577] [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: 12/14/2023] [Indexed: 12/19/2023] Open
Abstract
Sickle cell disease (SCD) is a genetic blood disorder that affects hemoglobin and increases stroke risk, particularly in childhood. This review examines the pathophysiological association between SCD and stroke, the classification of stroke types, risk factors, diagnosis, management, prevention, and prognosis. A comprehensive literature search was conducted via PubMed, Scopus, and Cochrane databases. Relevant studies on SCD and stroke pathophysiology, classification, epidemiology, diagnosis, treatment, and prevention were identified. Sickle cell disease causes red blood cells to become rigid and sickle-shaped, obstructing blood vessels. Recurrent sickling alters cerebral blood flow and damages vessel walls, often leading to ischemic or hemorrhagic strokes (HS). These occur most frequently in childhood, with ischemic strokes (IS) being more common. Key risk factors include a prior transient ischemic attack (TIA), low hemoglobin, and a high leukocyte count. Neuroimaging is essential for diagnosis and determining stroke type. Primary prevention centers on blood transfusions and hydroxyurea for those at high risk. Acute treatment involves promptly restoring blood flow and managing complications. However, significant knowledge gaps remain regarding stroke mechanisms, optimizing screening protocols, and improving long-term outcomes. This review synthesizes current evidence on SCD and stroke to highlight opportunities for further research and standardizing care protocols across institutions. Ultimately, a holistic perspective is critical for mitigating the high risk of debilitating strokes in this vulnerable patient population.
Collapse
Affiliation(s)
- Faisal Hakami
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Essam Alhazmi
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Wafa M Busayli
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | | | | | | | - Alyaj Hakami
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Renad A Othman
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Amal I Moafa
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | | | - Mohammed Ali Madkhali
- Internal Medicine, and Hematology and Oncology, Faculty of Medicine, Jazan University, Jazan, SAU
| |
Collapse
|
8
|
Silva M, Faustino P. From Stress to Sick(le) and Back Again-Oxidative/Antioxidant Mechanisms, Genetic Modulation, and Cerebrovascular Disease in Children with Sickle Cell Anemia. Antioxidants (Basel) 2023; 12:1977. [PMID: 38001830 PMCID: PMC10669666 DOI: 10.3390/antiox12111977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Sickle cell anemia (SCA) is a genetic disease caused by the homozygosity of the HBB:c.20A>T mutation, which results in the production of hemoglobin S (HbS). In hypoxic conditions, HbS suffers autoxidation and polymerizes inside red blood cells, altering their morphology into a sickle shape, with increased rigidity and fragility. This triggers complex pathophysiological mechanisms, including inflammation, cell adhesion, oxidative stress, and vaso-occlusion, along with metabolic alterations and endocrine complications. SCA is phenotypically heterogeneous due to the modulation of both environmental and genetic factors. Pediatric cerebrovascular disease (CVD), namely ischemic stroke and silent cerebral infarctions, is one of the most impactful manifestations. In this review, we highlight the role of oxidative stress in the pathophysiology of pediatric CVD. Since oxidative stress is an interdependent mechanism in vasculopathy, occurring alongside (or as result of) endothelial dysfunction, cell adhesion, inflammation, chronic hemolysis, ischemia-reperfusion injury, and vaso-occlusion, a brief overview of the main mechanisms involved is included. Moreover, the genetic modulation of CVD in SCA is discussed. The knowledge of the intricate network of altered mechanisms in SCA, and how it is affected by different genetic factors, is fundamental for the identification of potential therapeutic targets, drug development, and patient-specific treatment alternatives.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
- Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
9
|
De Clercq E. The magic of a methyl group: Biochemistry at the service of medicine. Biochem Pharmacol 2023; 216:115786. [PMID: 37660830 DOI: 10.1016/j.bcp.2023.115786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
A carbon-carbon linkage is created when a methyl group is implanted on dUMP, thus resulting in the formation of dTMP by thymidylate synthase. The methyl group is deleted by aromatase when androgens are converted to estrogens. The methyl group is rearranged with the help of vitamin B12 in the isomerization of methylmalonyl-CoA to succinyl-CoA. S-adenosylmethionine (SAM) serves as the universal methyl donor involved in the biosynthesis of adrenaline and creatine(phosphate). It also interferes with the 5'-mRNA capping and the degradation of catecholamines (i.e. adrenaline, noradrenaline). Cholesterol could be viewed as a conglomeration of methyl groups. Finally, as part of valine, two methyl functions participate in the origin of one of the most frequent hereditary diseases on earth, sickle cell anemia.
Collapse
Affiliation(s)
- Erik De Clercq
- Emeritus Professor at the KU Leuven (Belgium), Visiting Professor at the Jihočeská University of South Bohemia, České Budějovice, Czech Republic
| |
Collapse
|
10
|
Rogness VM, Juliette J, Khasabova IA, Gupta K, Khasabov SG, Simone DA. Descending Facilitation of Nociceptive Transmission From the Rostral Ventromedial Medulla Contributes to Hyperalgesia in Mice with Sickle Cell Disease. Neuroscience 2023; 526:1-12. [PMID: 37330194 PMCID: PMC10528639 DOI: 10.1016/j.neuroscience.2023.06.007] [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: 04/25/2023] [Revised: 05/26/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
Sickle cell disease (SCD) is an inherited blood disorder that is associated with acute episodic and chronic pain. Mice with SCD have robust hyperalgesia mediated, in part, by sensitization of spinal dorsal horn neurons. However, underlying mechanisms are not fully understood. Since the rostral ventromedial medulla (RVM) is a major component of descending circuitry that modulates nociceptive transmission in the spinal cord, we examined if the RVM contributes to hyperalgesia in mice with SCD. Injection of lidocaine, but not vehicle, into the RVM eliminated mechanical and heat hyperalgesia in sickle (HbSS-BERK) mice without altering mechanical and heat sensitivity in naïve C57B mice. These data indicate that the RVM contributes to the maintenance of hyperalgesia in mice with SCD. In electrophysiological studies, we determined the changes in response properties of RVM neurons that might contribute to hyperalgesia in sickle mice. Recordings were made from single ON, OFF, and Neutral cells in the RVM of sickle and control (HbAA-BERK) mice. Spontaneous activity and responses of ON, OFF and Neutral cells evoked by heat (50 °C) and mechanical (26 g) stimuli applied to the hind paw were compared between sickle and control mice. Although there were no differences in the proportions of functionally-identified neurons or spontaneous activity between sickle and control mice, evoked responses of ON cells to heat and mechanical stimuli were increased approximately 3-fold in sickle mice as compared to control mice. Thus, the RVM contributes to hyperalgesia in sickle mice via a specific ON cell-dependent descending facilitation of nociceptive transmission.
Collapse
Affiliation(s)
- Victoria M Rogness
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph Juliette
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Iryna A Khasabova
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kalpna Gupta
- Hematology/Oncology, Department of Medicine, University of California, Irvine and Southern California Institute for Research and Education, VA Medical Center, Long Beach, CA, USA
| | - Sergey G Khasabov
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Donald A Simone
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
11
|
Chambliss C, Stiles JK, Gee BE. Neuregulin-1 attenuates hemolysis- and ischemia induced-cerebrovascular inflammation associated with sickle cell disease. J Stroke Cerebrovasc Dis 2023; 32:106912. [PMID: 36473396 PMCID: PMC10448832 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/09/2022] Open
Abstract
OBJECTIVES Individuals with sickle cell disease (SCD) are at severely heightened risk for cerebrovascular injury and acute cerebrovascular events, including ischemic and hemorrhagic stroke, potentially leading to impaired development and life-long physical and cognitive disabilities. Cerebrovascular injury specific to SCD includes inflammation caused by underlying conditions of chronic hemolysis and reduced cerebrovascular perfusion. The objectives of this study were to investigate whether expression of neuregulin-1β (NRG-1), an endogenous neuroprotective polypeptide, is increased in SCD or experimental conditions mimicking the hemolysis and ischemic conditions of SCD, and to determine if treatment with exogenous NRG-1 reduces markers of cerebrovascular inflammation. MATERIALS AND METHODS Plasma and brain-specific NRG-1 levels were measured in transgenic SCD mice. Endogenous NRG-1 levels and response to experimental conditions of excess heme and ischemia were measured in cultured human brain microvascular cells and astrocytes. Pre-treatment with NRG-1 was used to determine NRG-1's ability to ameliorate resultant cerebrovascular inflammation. RESULTS Plasma and brain-specific NRG-1 were elevated in transgenic SCD mice compared to healthy controls. Neuregulin-1 expression was significantly increased in cultured human microvascular cells and astrocytes exposed to excess heme and ischemia. Pre-treatment with NRG-1 reduced inflammatory chemokine (CXCL-1 and CXCL-10) and adhesion molecule (ICAM-1 and VCAM-1) expression and increased pro-angiogenic factors (VEGF-A) in microvascular cells and astrocytes exposed to excess heme and ischemia. CONCLUSIONS Elevated NRG-1 in SCD is likely a protective endogenous response to ongoing cerebrovascular insults caused by chronic hemolysis and reduced cerebrovascular perfusion. Administration of NRG-1 to reduce cerebrovascular inflammation may be therapeutically beneficial in SCD and warrants continued investigation.
Collapse
Affiliation(s)
- Christopher Chambliss
- Pediatrics Institute, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Aflac Cancer and Blood Disorders Center, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Cardiovascular Research Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States.
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States.
| | - Beatrice E Gee
- Pediatrics Institute, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Aflac Cancer and Blood Disorders Center, 2015 Uppergate Drive, Atlanta, GA 30322, United States; Children's Healthcare of Atlanta, 35 Jesse Hill Jr Drive SE, Atlanta, GA 30303, United States; Department of Pediatrics, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310, United States
| |
Collapse
|
12
|
Cordovil K, Tassinari W, Oliveira RDVCD, Hökerberg Y. Social inequalities in the temporal trend of mortality from sickle cell disease in Brazil, 1996-2019. CAD SAUDE PUBLICA 2023; 39:e00256421. [PMID: 36651378 DOI: 10.1590/0102-311xen256421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 10/27/2022] [Indexed: 01/18/2023] Open
Abstract
Contrary to international trends, the mortality rate of sickle cell disease increased in Brazil after the implementation of the neonatal screening program, probably due to improving access to diagnosis. This study aimed to assess differences in the temporal trend of the mortality rate and median age at death from sickle cell disease in Brazil, considering implemented measures to expand diagnosis, and improve health care access in-country and in the international scenario. Time series were extracted from the Brazilian Mortality Information System from 1996 to 2019. Changes in the mortality rate and median age at death were verified via segmented regression models, which were stratified by sex, region of residence, and age. Most deaths occurred in non-white people, young adults, and the Southeast and Northeast population. Sickle cell disease mortality rate increased until 2010 (13.31%; 95%CI: 6.37; 20.70), particularly in individuals aged 30 years or more (12.78%; 95%CI: 2.98; 23.53) and in the Northeast (12.27%; 95%CI: 8.92; 15.72). Most deaths occurred in the second decade of life (3.01 deaths/million), with a 59% increase in the median age of death in Brazil, from 27.6 to 30.3 years, more pronounced in females and the North Region. The observed gain in the survival of sickle cell disease in Brazil is still much lower than in developed countries and presents regional disparities, probably due to the lack of access to health care and recent treatments, such as hydroxyurea, still restricted to hematological referral centers in Brazilian capitals.
Collapse
Affiliation(s)
- Karen Cordovil
- Escola Nacional de Saúde Pública Sergio Arouca, Fundação Oswaldo Cruz, Rio de Janeiro, Brasil
| | - Wagner Tassinari
- Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brasil
| | | | - Yara Hökerberg
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brasil
| |
Collapse
|
13
|
Viswanathan VK, Ramanan SP, Beale J, Subramanian S, Mounasamy V, Sambandam S. How does sickle cell disease affect the peri-operative outcome in patients undergoing total knee arthroplasty? A large-scale, National Inpatient Sample-based study. Arch Orthop Trauma Surg 2023:10.1007/s00402-022-04762-1. [PMID: 36592196 DOI: 10.1007/s00402-022-04762-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
INTRODUCTION In view of the vaso-occlusive pathophysiology affecting osseous micro-circulation, sickle cell disease (SCD) is well known to present with diverse skeletal and arthritic manifestations. With prolonged life-expectancy over the past decades, there has been a progressive increase in the proportion of SCD patients requiring joint reconstructions. Owing to the paucity of evidence in the literature, the post-operative complication rates and outcome in these patients following total knee arthroplasty (TKA) are still largely unknown. METHODS Based on the National Inpatient Sample (NIS) database (using ICD-10 CMP code), patients who underwent TKA between 2016 and 2019 were identified. The cohort were classified into two groups: A-those with SCD; and B-those without. The data on patients' demographics, co-morbidities, details regarding hospital stay including expenditure incurred, and complications were analyzed and compared. RESULTS Overall, 558,361 patients underwent unilateral, primary TKA; among whom, 493 (0.1%) were known cases of SCD (group A). Group A included a significantly greater proportion of younger (60.14 ± 10.87 vs 66.72 ± 9.50 years; p < 0.001), male (77.3 vs 61.5%; p < 0.001); and African-American (88.2 vs 8.3%B; p < 0.001) patients, in comparison with group B. Group A patients were also at a significantly higher risk for longer duration of peri-operative hospital stay (p < 0.001), greater health-care costs incurred (p < 0.001), and greater need for alternative step-down health-care facilities (p < 0.001) following discharge. Among the SCD patients, 24.7%, 20.9% and 24.9% developed acute chest syndrome, pain crisis and splenic sequestration crisis, respectively during the peri-operative period. Group A patients had a statistically greater incidence of acute renal failure (ARF; p = 0.014), need for blood transfusion (p < 0.001) and deep vein thrombosis (DVT; p = 0.03) during the early admission period. CONCLUSION The presence of SCD substantially lengthens the duration of hospital stay and enhances health care-associated expenditure in patients undergoing TKA. SCD patients are at significantly higher risk for systemic complications including acute chest syndrome, pain crisis, splenic sequestration crisis, acute renal failure, higher need for blood transfusions and deep venous thrombosis during the initial peri-operative period following TKA.
Collapse
Affiliation(s)
| | | | - Jack Beale
- Department of Orthopedics, University of Texas Southwestern, Dallas, TX, USA
| | - Surabhi Subramanian
- Pediatric Radiology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Varatharaj Mounasamy
- Chief of Orthopedics, Department of Orthopedics, University of Texas Southwestern, Dallas VAMC, Dallas, TX, USA
| | | |
Collapse
|
14
|
Kour D, Ali M, Khajuria P, Sharma K, Ghosh P, Kaur S, Mahajan S, Ramajayan P, Bharate SS, Bhardwaj S, Sawant SD, Reddy DS, Kumar A. Flurbiprofen inhibits heme induced NLRP3 inflammasome in Berkeley sickle cell disease mice. Front Pharmacol 2023; 14:1123734. [PMID: 37180702 PMCID: PMC10171431 DOI: 10.3389/fphar.2023.1123734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Sickle cell disease (SCD) is accompanied by several complications, which emanate from the sickling of erythrocytes due to a point mutation in the β-globin chain of hemoglobin. Sickled erythrocytes are unable to move smoothly through small blood capillaries and therefore, cause vaso occlusion and severe pain. Apart from pain, continuous lysis of fragile sickled erythrocytes leads to the release of heme, which is a strong activator of the NLRP3 inflammasome, thus producing chronic inflammation in sickle cell disease. In this study, we identified flurbiprofen among other COX-2 inhibitors to be a potent inhibitor of heme-induced NLRP3 inflammasome. We found that apart from being a nociceptive agent, flurbiprofen exerts a strong anti-inflammatory effect by suppressing NF-κB signaling, which was evidenced by reduced levels of TNF-α and IL-6 in wild-type and sickle cell disease Berkeley mice models. Our data further demonstrated the protective effect of flurbiprofen on liver, lungs, and spleen in Berkeley mice. The current sickle cell disease pain management regime relies mainly on opiate drugs, which is accompanied by several side effects without modifying the sickle cell disease-related pathology. Considering the potent role of flurbiprofen in inhibiting NLRP3 inflammasome and other inflammatory cytokines in sickle cell disease, our data suggests that it can be explored further for better sickle cell disease pain management along with the possibility of disease modification.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Mehboob Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Palash Ghosh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sukhleen Kaur
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Surbhi Mahajan
- Department of Pathology, Government Medical College, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Sonali S. Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai, India
| | - Subhash Bhardwaj
- Department of Pathology, Government Medical College, Jammu, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - D. Srinivasa Reddy
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| |
Collapse
|
15
|
Xu JZ, Conrey A, Frey I, Gwaabe E, Menapace LA, Tumburu L, Lundt M, Lequang T, Li Q, Glass K, Dunkelberger EB, Iyer V, Mangus H, Kung C, Dang L, Kosinski PA, Hawkins P, Jeffries N, Eaton WA, Lay Thein S. A phase 1 dose escalation study of the pyruvate kinase activator mitapivat (AG-348) in sickle cell disease. Blood 2022; 140:2053-2062. [PMID: 35576529 PMCID: PMC9837441 DOI: 10.1182/blood.2022015403] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/11/2022] [Indexed: 01/21/2023] Open
Abstract
Polymerization of deoxygenated hemoglobin S underlies the pathophysiology of sickle cell disease (SCD). In activating red blood cell pyruvate kinase and glycolysis, mitapivat (AG-348) increases adenosine triphosphate (ATP) levels and decreases the 2,3-diphosphoglycerate (2,3-DPG) concentration, an upstream precursor in glycolysis. Both changes have therapeutic potential for patients with SCD. Here, we evaluated the safety and tolerability of multiple ascending doses of mitapivat in adults with SCD with no recent blood transfusions or changes in hydroxyurea or l-glutamine therapy. Seventeen subjects were enrolled; 1 subject was withdrawn shortly after starting the study. Sixteen subjects completed 3 ascending dose levels of mitapivat (5, 20, and 50 mg, twice daily [BID]) for 2 weeks each; following a protocol amendment, the dose was escalated to 100 mg BID in 9 subjects. Mitapivat was well tolerated at all dose levels, with the most common treatment-emergent adverse events (AEs) being insomnia, headache, and hypertension. Six serious AEs (SAEs) included 4 vaso-occlusive crises (VOCs), non-VOC-related shoulder pain, and a preexisting pulmonary embolism. Two VOCs occurred during drug taper and were possibly drug related; no other SAEs were drug related. Mean hemoglobin increase at the 50 mg BID dose level was 1.2 g/dL, with 9 of 16 (56.3%) patients achieving a hemoglobin response of a ≥1 g/dL increase compared with baseline. Mean reductions in hemolytic markers and dose-dependent decreases in 2,3-DPG and increases in ATP were also observed. This study provides proof of concept that mitapivat has disease-modifying potential in patients with SCD. This trial was registered at www.clinicaltrials.gov as #NCT04000165.
Collapse
Affiliation(s)
- Julia Z. Xu
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Anna Conrey
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ingrid Frey
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Eveline Gwaabe
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Laurel A. Menapace
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Laxminath Tumburu
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Maureen Lundt
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Timothy Lequang
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Quan Li
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Kristen Glass
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Emily B. Dunkelberger
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | | | | | | - Lenny Dang
- Agios Pharmaceuticals, Inc., Cambridge, MA
| | | | | | - Neal Jeffries
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - William A. Eaton
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
16
|
Design, Synthesis, and Investigation of Novel Nitric Oxide (NO)-Releasing Aromatic Aldehyde as Drug Candidates for the Treatment of Sickle Cell Disease. Molecules 2022; 27:molecules27206835. [PMID: 36296435 PMCID: PMC9610770 DOI: 10.3390/molecules27206835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/29/2022] Open
Abstract
Sickle cell disease (SCD) is caused by a single-point mutation, and the ensuing deoxygenation-induced polymerization of sickle hemoglobin (HbS), and reduction in bioavailability of vascular nitric oxide (NO), contribute to the pathogenesis of the disease. In a proof-of-concept study, we successfully incorporated nitrate ester groups onto two previously studied potent antisickling aromatic aldehydes, TD7 and VZHE039, to form TD7-NO and VZHE039-NO hybrids, respectively. These compounds are stable in buffer but demonstrated the expected release of NO in whole blood in vitro and in mice. The more promising VZHE039-NO retained the functional and antisickling activities of the parent VZHE039 molecule. Moreover, VZHE039-NO, unlike VZHE039, significantly attenuated RBC adhesion to laminin, suggesting this compound has potential in vivo RBC anti-adhesion properties relevant to vaso-occlusive events. Crystallographic studies show that, as with VZHE039, VZHE039-NO also binds to liganded Hb to make similar protein interactions. The knowledge gained during these investigations provides a unique opportunity to generate a superior candidate drug in SCD with enhanced benefits.
Collapse
|
17
|
Revisiting anemia in sickle cell disease and finding the balance with therapeutic approaches. Blood 2022; 139:3030-3039. [PMID: 35587865 DOI: 10.1182/blood.2021013873] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/05/2021] [Indexed: 11/20/2022] Open
Abstract
Chronic hemolytic anemia and intermittent acute pain episodes are the 2 hallmark characteristics of sickle cell disease (SCD). Anemia in SCD not only signals a reduction of red cell mass and oxygen delivery, but also ongoing red cell breakdown and release of cell-free hemoglobin, which together contribute to a number of pathophysiological responses and play a key role in the pathogenesis of cumulative multiorgan damage. However, although anemia is clearly associated with many detrimental outcomes, it may also have an advantage in SCD in lowering risks of potential viscosity-related complications. Until recently, clinical drug development for SCD has predominantly targeted a reduction in the frequency of vaso-occlusive crises as an endpoint, but increasingly, more attention is being directed toward addressing the contribution of chronic anemia to poor outcomes in SCD. This article aims to explore the complex pathophysiology and mechanisms of anemia in SCD, as well as the need to balance the benefits of raising hemoglobin levels with the potential risks of increasing blood viscosity, in the context of the current therapeutic landscape for anemia in SCD.
Collapse
|
18
|
Alhashimi RT, Ghatge MS, Donkor AK, Deshpande TM, Anabaraonye N, Alramadhani D, Danso-Danquah R, Huang B, Zhang Y, Musayev FN, Abdulmalik O, Safo MK. Design, Synthesis, and Antisickling Investigation of a Nitric Oxide-Releasing Prodrug of 5HMF for the Treatment of Sickle Cell Disease. Biomolecules 2022; 12:696. [PMID: 35625623 PMCID: PMC9138457 DOI: 10.3390/biom12050696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/02/2023] Open
Abstract
5-hydroxyfurfural (5HMF), an allosteric effector of hemoglobin (Hb) with an ability to increase Hb affinity for oxygen has been studied extensively for its antisickling effect in vitro and in vivo, and in humans for the treatment of sickle cell disease (SCD). One of the downstream pathophysiologies of SCD is nitric oxide (NO) deficiency, therefore increasing NO (bio)availability is known to mitigate the severity of SCD symptoms. We report the synthesis of an NO-releasing prodrug of 5HMF (5HMF-NO), which in vivo, is expected to be bio-transformed into 5HMF and NO, with concomitant therapeutic activities. In vitro studies showed that when incubated with whole blood, 5HMF-NO releases NO, as anticipated. When incubated with sickle blood, 5HMF-NO formed Schiff base adduct with Hb, increased Hb affinity for oxygen, and prevented hypoxia-induced erythrocyte sickling, which at 1 mM concentration were 16%, 10% and 27%, respectively, compared to 21%, 18% and 21% for 5HMF. Crystal structures of 5HMF-NO with Hb showed 5HMF-NO bound to unliganded (deoxygenated) Hb, while the hydrolyzed product, 5HMF bound to liganded (carbonmonoxy-ligated) Hb. Our findings from this proof-of-concept study suggest that the incorporation of NO donor group to 5HMF and analogous molecules could be a novel beneficial strategy to treat SCD and warrants further detailed in vivo studies.
Collapse
Affiliation(s)
- Rana T. Alhashimi
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Mohini S. Ghatge
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Akua K. Donkor
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | | | - Nancy Anabaraonye
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (N.A.); (O.A.)
| | - Dina Alramadhani
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Richmond Danso-Danquah
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Boshi Huang
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Yan Zhang
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Faik N. Musayev
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| | - Osheiza Abdulmalik
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; (N.A.); (O.A.)
| | - Martin K. Safo
- Department of Medicinal Chemistry and The Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (R.T.A.); (M.S.G.); (A.K.D.); (D.A.); (R.D.-D.); (B.H.); (Y.Z.); (F.N.M.)
| |
Collapse
|
19
|
Ratanasopa K, Bulow L. Calorimetric Characterisation of the Binding Reaction Between Human Ferric Haemoglobins and Haptoglobin to Develop a Drug for Removal of Cell-Free Haemoglobin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1395:341-345. [PMID: 36527659 DOI: 10.1007/978-3-031-14190-4_55] [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: 12/23/2022]
Abstract
High levels of cell-free haemoglobin (Hb) may occur in plasma as a consequence of e.g., pathological haemolysis or blood transfusion. These Hb molecules can be removed from blood circulation by forming a complex with the acute-phase protein haptoglobin (Hp) and thereby can also the intrinsic toxicity of free Hb be limited. In this study it is shown that ferric HbA, HbF, HbE and HbS, respectively, all bind firmly to Hp at 25 °C. By using isothermal titration calorimetry (ITC), it is demonstrated that ferric HbF has higher affinity to Hp (Ka = 2.79 ± 0.29 ×109 M-1) compared with HbA and HbS (1.91 ± 0.24 ×109 M-1) and 1.41 ± 0.34 ×109 M-1 for HbA and HbS, respectively. In addition, the affinity constant for HbE is slightly lower than the other haemoglobins (0.47 ± 0.40 ×109 M-1). Since Hp shows a general and high affinity to all Hb variants tested, it can be concluded that Hp may be useful as a therapeutic agent for several different haemolytic conditions by intravenous injection.
Collapse
Affiliation(s)
| | - Leif Bulow
- Department of Chemistry, Pure and Applied Biochemistry, Lund University, Lund, Sweden.
| |
Collapse
|
20
|
Lin Z, Lance E, McIntyre T, Li Y, Liu P, Lim C, Fan H, Tekes A, Cannon A, Casella JF, Lu H. Imaging Blood-Brain Barrier Permeability Through MRI in Pediatric Sickle Cell Disease: A Feasibility Study. J Magn Reson Imaging 2021; 55:1551-1558. [PMID: 34676938 PMCID: PMC9018466 DOI: 10.1002/jmri.27965] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption may lead to endothelium dysfunction and inflammation in sickle cell disease (SCD). However, abnormalities of BBB in SCD, especially in pediatric patients for whom contrast agent administration less than optimal, have not been fully characterized. PURPOSE To examine BBB permeability to water in a group of pediatric SCD participants using a non-invasive magnetic resonance imaging technique. We hypothesized that SCD participants will have increased BBB permeability. STUDY TYPE Prospective cross-sectional. POPULATION Twenty-six pediatric participants (10 ± 1 years, 15F/11M) were enrolled, including 21 SCD participants and 5 sickle cell trait (SCT) participants, who were siblings of SCD patients. FIELD STRENGTH/SEQUENCE 3 T. Water extraction with phase-contrast arterial spin tagging with echo-planer imaging, phase-contrast and T1 -weighted magnetization-prepared rapid acquisition of gradient echo. ASSESSMENT Water extraction fraction (E), BBB permeability-surface area product (PS), cerebral blood flow, hematological measures (hemoglobin, hematocrit, hemoglobin S), neuropsychological scores (including domains of intellectual ability, attention and executive function, academic achievement and adaptive function, and a composite score). Regions of interest were drawn by Z.L. (6 years of experience). STATISTICAL TESTS Wilcoxon rank sum test and chi-square test for group comparison of demographics. Multiple linear regression analysis of PS with diagnostic category (SCD or SCT), hematological measures, and neuropsychological scores. A two-tailed P value of 0.05 or less was considered statistically significant. RESULTS Compared with SCT participants, SCD participants had a significantly higher BBB permeability to water (SCD: 207.0 ± 33.3 mL/100 g/minute, SCT: 171.2 ± 27.2 mL/100 g/minute). SCD participants with typically more severe phenotypes also had a significantly leakier BBB than those with typically milder phenotypes (severe: 217.3 ± 31.7 mL/100 g/minute, mild: 193.3 ± 31.8 mL/100 g/minute). Furthermore, more severe BBB disruption was associated with worse hematological symptoms, including lower hemoglobin concentrations (β = -8.84, 95% confidence interval [CI] [-14.69, -3.00]), lower hematocrits (β = -2.96, 95% CI [-4.84, -1.08]), and higher hemoglobin S fraction (β = 0.77, 95% CI [0.014, 1.53]). DATA CONCLUSION These findings support a potential role for BBB dysfunction in SCD pathogenesis of ischemic injury. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Zixuan Lin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eboni Lance
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tiffany McIntyre
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Yang Li
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peiying Liu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chantelle Lim
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hongli Fan
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aylin Tekes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alicia Cannon
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - James F Casella
- Department of Pediatrics, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanzhang Lu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Moussa O, Chen RWS. Hemoglobinopathies: ocular manifestations in children and adolescents. Ther Adv Ophthalmol 2021; 13:25158414211022882. [PMID: 34263135 PMCID: PMC8252354 DOI: 10.1177/25158414211022882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022] Open
Abstract
Hemoglobinopathies are genetic disorders that lead to abnormal structure of the hemoglobin molecule. Sickle cell disease, the most common inherited blood disorder, is characterized by defective oxygen transport. Almost every part of the eye can be affected by sickle cell disease; however, proliferative sickle cell retinopathy is the primary cause of vision loss, either from vitreous hemorrhage or retinal detachment. Here we review the various manifestations of hemoglobinopathies on the eyes of children and adolescents, with a specific focus on sickle cell disease and its different phenotypes. Newer, more sensitive ophthalmological imaging modalities, including ultra-widefield fluorescein angiography, spectral-domain optical coherence tomography, and optical coherence tomography angiography, are available. These sensitive modalities allow for a more thorough examination of the retinal periphery where sickle cell retinopathy is often present. Utilization of such modalities will help with the early detection of the disease in children, which provide a better understanding of the pathogenesis of the disease and guide future screening and treatment regimens.
Collapse
Affiliation(s)
- Omar Moussa
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA
| | - Royce W S Chen
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, 635 W. 165th St., New York, NY 10032, USA
| |
Collapse
|
22
|
Juttukonda MR, Donahue MJ, Waddle SL, Davis LT, Lee CA, Patel NJ, Pruthi S, Kassim AA, Jordan LC. Reduced oxygen extraction efficiency in sickle cell anemia patients with evidence of cerebral capillary shunting. J Cereb Blood Flow Metab 2021; 41:546-560. [PMID: 32281458 PMCID: PMC7922746 DOI: 10.1177/0271678x20913123] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arterial spin labeling (ASL) magnetic resonance imaging (MRI) utilizes arterial blood water as an endogenous contrast agent to provide a quantitative measure of cerebral blood flow (CBF). Recently, hyperintense signal within dural venous sinuses in ASL images of sickle cell anemia (SCA) patients has been shown to be consistent with elevated flow velocities and may indicate capillary shunting and reduced oxygen extraction. Here, we performed oxygen extraction fraction (OEF) and CBF measurements in adults (cumulative n = 114) with (n = 69) and without (n = 45) SCA to test the hypothesis that hyperintense venous ASL signal is associated with reduced OEF. Higher categorical scores of shunting on ASL MRI were associated with lower OEF in participants with silent cerebral infarcts or white matter hyperintensities (p = 0.003), but not in those without lesions (p = 0.551). These findings indicate that venous hyperintense signal in ASL images in SCA patients may represent a marker of capillary-level disturbances in oxygen exchange efficiency and small vessel pathology.
Collapse
Affiliation(s)
- Meher R Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Spencer L Waddle
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry T Davis
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chelsea A Lee
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Niral J Patel
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sumit Pruthi
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adetola A Kassim
- Department of Medicine, Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lori C Jordan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
23
|
Characterization of microvascular disease in pediatric sickle cell disease using nailfold capillaroscopy. Microvasc Res 2021; 136:104150. [PMID: 33647341 DOI: 10.1016/j.mvr.2021.104150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 11/22/2022]
Abstract
Sickle cell disease (SCD) is a disorder with repetitive vaso-occlusive crises resulting in microvascular obstruction and tissue ischemia that may lead to multi-organ ischemia and dysfunction. Nailfold videocapillaroscopy (NFC) is an imaging technique utilized in clinical rheumatology to visualize capillaries located near the fingertip. To characterize NFC abnormalities in the setting of pediatric SCD, we performed NFC using a video capillaroscope on 8 digits in 44 stable SCD patients and 65 age matched healthy controls. Mean capillary number was lower (6.4 ± 1.3 vs 7.5 ± 1.8, p = 0.001) in the SCD group compared to controls. The percentage of dilated capillaries was similar (7.1 ± 8.3 vs. 5.9 ± 8.2, p = 0.4). The large majority of capillaries visualized in the SCD and control groups were normal capillary types per the EULAR definition, with a similar percentage of normal, nonspecific capillary morphologies and abnormal types. Regarding normal capillary sub-types, the SCD group and controls exhibited similar percentages of stereotype hairpin shapes, and tortuous or once or twice crossing type capillaries. On multivariate analyses, mean capillary number was independently associated with SCD after adjusting for age, body mass index, systolic blood pressure and gender. In conclusion, pediatric SCD is associated with lower capillary number but similar percentage of dilated capillaries and morphology on NFC. In our SCD cohort, capillary number was unrelated to our available markers of disease severity, including history of sickle crises, previous hospitalization for crises or Hemoglobin F levels.
Collapse
|
24
|
Ingerski LM, Loew M, Porter JS, Su Y, Zhang H, Hankins JS, Wang WC. Use of Wise Device Technology to Measure Adherence to Hydroxyurea Therapy in Youth With Sickle Cell Disease. J Pediatr Hematol Oncol 2021; 43:e19-e25. [PMID: 33235145 DOI: 10.1097/mph.0000000000001997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Despite broad support for hydroxyurea (HU) therapy, suboptimal adherence is reported for youth with sickle cell disease. Valid adherence measurement is crucial to understanding the relationship between medication behavior, disease response, and patient-centered health outcomes. The current pilot study examined the feasibility of the Wise electronic device for longitudinal HU adherence measurement in a sample of 36 youths prescribed HU. The study also explored the association between HU adherence, as measured by the Wise device, with other adherence measures (ie, family report, lab values, pill count, and medication possession ratio). A measure of family-reported acceptability was also completed. Overall, results supported the feasibility of the Wise device (rate of consent=82%, device use=75%, device failure=3%) for HU adherence measurement and most families rated their experience using their device positively (favorable responses ranged from 67% to 100%). Associations between HU adherence, as measured by the Wise device, and other adherence measures were not significant. Overall, the feasibility was supported. The Wise device allows longitudinal measurement of adherence with HU from initiation as a young child (ie, with liquid formulations) through adolescence and provides a novel means of adherence measurement for both clinical and research use.
Collapse
Affiliation(s)
| | | | | | | | | | - Jane S Hankins
- Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Winfred C Wang
- Hematology, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
25
|
Yuan S, Jordan LC, Davis LT, Cogswell PM, Lee CA, Patel NJ, Waddle SL, Juttukonda M, Sky Jones R, Griffin A, Donahue MJ. A cross-sectional, case-control study of intracranial arterial wall thickness and complete blood count measures in sickle cell disease. Br J Haematol 2020; 192:769-777. [PMID: 33326595 DOI: 10.1111/bjh.17262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/25/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
In sickle cell disease (SCD), cerebral oxygen delivery is dependent on the cerebral vasculature's ability to increase blood flow and volume through relaxation of the smooth muscle that lines intracranial arteries. We hypothesised that anaemia extent and/or circulating markers of inflammation lead to concentric macrovascular arterial wall thickening, visible on intracranial vessel wall magnetic resonance imaging (VW-MRI). Adult and pediatric SCD (n = 69; age = 19.9 ± 8.6 years) participants and age- and sex-matched control participants (n = 38; age = 22.2 ± 8.9 years) underwent 3-Tesla VW-MRI; two raters measured basilar and bilateral supraclinoid internal carotid artery (ICA) wall thickness independently. Mean wall thickness was compared with demographic, cerebrovascular and haematological variables. Mean vessel wall thickness was elevated (P < 0·001) in SCD (1·07 ± 0·19 mm) compared to controls (0·97 ± 0·07 mm) after controlling for age and sex. Vessel wall thickness was higher in participants on chronic transfusions (P = 0·013). No significant relationship between vessel wall thickness and flow velocity, haematocrit, white blood cell count or platelet count was observed; however, trends (P < 0·10) for wall thickness increasing with decreasing haematocrit and increasing white blood cell count were noted. Findings are discussed in the context of how anaemia and circulating inflammatory markers may impact arterial wall morphology.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lori C Jordan
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry T Davis
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Petrice M Cogswell
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Chelsea A Lee
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Niral J Patel
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Spencer L Waddle
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meher Juttukonda
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - R Sky Jones
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison Griffin
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
26
|
Argueta DA, Aich A, Muqolli F, Cherukury H, Sagi V, DiPatrizio NV, Gupta K. Considerations for Cannabis Use to Treat Pain in Sickle Cell Disease. J Clin Med 2020; 9:E3902. [PMID: 33271850 PMCID: PMC7761429 DOI: 10.3390/jcm9123902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 12/18/2022] Open
Abstract
Pain in Sickle Cell Disease (SCD) is a major comorbidity and unique with acute pain due to recurrent and episodic vaso-occlusive crises as well as chronic pain, which can span an individual's entire life. Opioids are the mainstay treatment for pain in SCD. Due to recent health crises raised by adverse effects including deaths from opioid use, pain management in SCD is adversely affected. Cannabis and its products are most widely used for pain in multiple conditions and also by patients with SCD on their own. With the availability of "Medical Cannabis" and approval to use cannabis as medicine across majority of States in the United States as well as over-the-counter preparations, cannabis products are being used increasingly for SCD. The reliability of many of these products remains questionable, which poses a major health risk to the vulnerable individuals seeking pain relief. Therefore, this review provides up to date insights into available categories of cannabis-based treatment strategies, their mechanism of action and pre-clinical and clinical outcomes in SCD. It provides evidence for the benefits and risks of cannabis use in SCD and cautions about the unreliable and unvalidated products that may be adulterated with life-threatening non-cannabis compounds.
Collapse
Affiliation(s)
- Donovan A. Argueta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92868, USA; (D.A.A.); (A.A.); (F.M.); (H.C.)
| | - Anupam Aich
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92868, USA; (D.A.A.); (A.A.); (F.M.); (H.C.)
| | - Fjolla Muqolli
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92868, USA; (D.A.A.); (A.A.); (F.M.); (H.C.)
| | - Hemanth Cherukury
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92868, USA; (D.A.A.); (A.A.); (F.M.); (H.C.)
| | - Varun Sagi
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Twin Cities, MN 55455, USA;
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA;
| | - Kalpna Gupta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, CA 92868, USA; (D.A.A.); (A.A.); (F.M.); (H.C.)
- Southern California Institute for Research and Education, Long Beach VA Medical Center, Long Beach, CA 90822, USA
| |
Collapse
|
27
|
Veluswamy S, Shah P, Khaleel M, Thuptimdang W, Chalacheva P, Sunwoo J, Denton CC, Kato R, Detterich J, Wood JC, Sposto R, Khoo MCK, Zeltzer L, Coates TD. Progressive vasoconstriction with sequential thermal stimulation indicates vascular dysautonomia in sickle cell disease. Blood 2020; 136:1191-1200. [PMID: 32518948 PMCID: PMC7472716 DOI: 10.1182/blood.2020005045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Persons with sickle cell disease (SCD) exhibit subjective hypersensitivity to cold and heat perception in experimental settings, and triggers such as cold exposure are known to precipitate vaso-occlusive crises by still unclear mechanisms. Decreased microvascular blood flow (MBF) increases the likelihood of vaso-occlusion by increasing entrapment of sickled red blood cells in the microvasculature. Because those with SCD have dysautonomia, we anticipated that thermal exposure would induce autonomic hypersensitivity of their microvasculature with an increased propensity toward vasoconstriction. We exposed 17 patients with SCD and 16 control participants to a sequence of predetermined threshold temperatures for cold and heat detection and cold and heat pain via a thermode placed on the right hand. MBF was measured on the contralateral hand by photoplethysmography, and cardiac autonomic balance was assessed by determining heart rate variability. Thermal stimuli at both detection and pain thresholds caused a significant decrease in MBF in the contralateral hand within seconds of stimulus application, with patients with SCD showing significantly stronger vasoconstriction (P = .019). Furthermore, patients with SCD showed a greater progressive decrease in blood flow than did the controls, with poor recovery between episodes of thermal stimulation (P = .042). They had faster vasoconstriction than the controls (P = .033), especially with cold detection stimulus. Individuals with higher anxiety also experienced more rapid vasoconstriction (P = .007). Augmented vasoconstriction responses and progressive decreases in perfusion with repeated thermal stimulation in SCD are indicative of autonomic hypersensitivity in the microvasculature. These effects are likely to increase red cell entrapment in response to clinical triggers such as cold or stress, which have been associated with vaso-occlusive crises in SCD.
Collapse
Affiliation(s)
- Saranya Veluswamy
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA
| | - Payal Shah
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA
| | - Maha Khaleel
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA
| | - Wanwara Thuptimdang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Patjanaporn Chalacheva
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - John Sunwoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Christopher C Denton
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA
| | | | | | | | - Richard Sposto
- Division of Preventive Medicine, Children's Hospital Los Angeles, Los Angeles, CA; and
| | - Michael C K Khoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Lonnie Zeltzer
- Pediatric Pain Program, University of California, Los Angeles, CA
| | - Thomas D Coates
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA
| |
Collapse
|
28
|
Gilyard SN, Hamlin SL, Johnson JO, Herr KD. Imaging review of sickle cell disease for the emergency radiologist. Emerg Radiol 2020; 28:153-164. [PMID: 32734483 DOI: 10.1007/s10140-020-01828-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
Abstract
Sickle cell disease (SCD), one of the most common inherited genetic syndromes in the USA, is characterized by recurring episodes of acute illness and progressive multisystem organ injury. Individuals with SCD frequently present to the emergency department for a spectrum of complications, such as vaso-occlusive crises, infection, cholecystitis, and stroke. Imaging correlates for most of these presentations exist, positioning the emergency radiologist to play a pivotal role in facilitating patient care. Using a systems-based approach, we describe the acute and chronic imaging manifestations of SCD that an emergency radiologist can expect to encounter in most practice settings, highlighting the unique pathophysiology of this disorder that typically underlies the imaging findings.
Collapse
Affiliation(s)
- Shenise N Gilyard
- Emory University School of Medicine, Atlanta, USA. .,Division of Emergency and Trauma Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, 1364 Clifton Road NE Suite #D112, Atlanta, USA.
| | - Scott L Hamlin
- Emory University School of Medicine, Atlanta, USA.,Division of Emergency and Trauma Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, 1364 Clifton Road NE Suite #D112, Atlanta, USA
| | - Jamlik-Omari Johnson
- Emory University School of Medicine, Atlanta, USA.,Division of Emergency and Trauma Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, 1364 Clifton Road NE Suite #D112, Atlanta, USA
| | - Keith D Herr
- Emory University School of Medicine, Atlanta, USA.,Division of Emergency and Trauma Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, 1364 Clifton Road NE Suite #D112, Atlanta, USA
| |
Collapse
|
29
|
Martí-Carvajal AJ, Martí-Amarista CE. Interventions for treating intrahepatic cholestasis in people with sickle cell disease. Cochrane Database Syst Rev 2020; 6:CD010985. [PMID: 32567054 PMCID: PMC7388850 DOI: 10.1002/14651858.cd010985.pub4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Sickle cell disease is the most common hemoglobinopathy occurring worldwide and sickle cell intrahepatic cholestasis is a complication long recognized in this population. Cholestatic liver diseases are characterized by impaired formation or excretion (or both) of bile from the liver. There is a need to assess the clinical benefits and harms of the interventions used to treat intrahepatic cholestasis in people with sickle cell disease. This is an update of a previously published Cochrane Review. OBJECTIVES To assess the benefits and harms of the interventions for treating intrahepatic cholestasis in people with sickle cell disease. SEARCH METHODS We searched the Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register, which comprises references identified from comprehensive electronic database searches and handsearching of relevant journals and abstract books of conference proceedings. We also searched the LILACS database (1982 to 21 January 2020), the WHO International Clinical Trials Registry Platform Search Portal and ClinicalTrials.gov (21 January 2020). Date of last search of the Cochrane Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register: 25 November 2019. SELECTION CRITERIA We searched for published or unpublished randomised controlled trials. DATA COLLECTION AND ANALYSIS Each author intended to independently extract data, assess the risk of bias of the trials by standard Cochrane methodologies and assess the quality of the evidence using the GRADE criteria; however, no trials were included in the review. MAIN RESULTS We did not identify any randomised controlled trials. AUTHORS' CONCLUSIONS This updated Cochrane Review did not identify any randomised controlled trials assessing interventions for treating intrahepatic cholestasis in people with sickle cell disease. Randomised controlled trials are needed to establish the optimum treatment for this condition.
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE (Cochrane Ecuador), Quito, Ecuador
- School of Medicine, Universidad Francisco de Vitoria (Cochrane Madrid), Madrid, Spain
| | | |
Collapse
|
30
|
Abstract
STUDY DESIGN A retrospective study. OBJECTIVE To determine the impact of sickle cell anemia on perioperative outcomes and resource utilization in elective spinal fusion surgery. SUMMARY OF BACKGROUND DATA Sickle cell anemia has been identified as an important surgical risk factor in otolaryngology, cardiothoracic surgery, general surgery, and total joint arthroplasty. However, the impact of sickle cell anemia on elective spine surgery is unknown. METHODS Hospitalizations for elective spinal fusion surgery between the years of 2001-2014 from the US National Inpatient Sample were identified using ICD-9-CM codes and patients were grouped into those with and without sickle cell anemia. The main outcome measures were in-hospital neurological, respiratory, cardiac, gastrointestinal, renal and urinary, pulmonary embolism, and wound-related complications and mortality. Length of stay and inpatient costs were also collected. Multivariable logistic regressions were conducted to compare the in-hospital outcomes of patients undergoing elective spinal fusion with or without sickle cell anemia. RESULTS From a total of 4,542,719 patients undergoing elective spinal fusions from 2001 to 2014, 456 sickle cell disease patients were identified. Sickle cell anemia is a significant independent predictor for pulmonary embolism [odds ratio (OR)=7.37; confidence interval (CI), 4.27-12.71; P<0.001], respiratory complications (OR=2.36; CI, 1.63-3.42; P<0.001), wound complications (OR=3.84; CI, 2.72-5.44; P<0.001), and overall inpatient complications (OR=2.58; CI, 2.05-3.25; P<0.001). Sickle cell anemia patients also have significantly longer length of stay (7.0 vs. 3.8 d; P<0.001) and higher inpatient costs ($20,794 vs. $17,608 P<0.05). CONCLUSIONS Sickle cell anemia is associated with increased risk of perioperative complications and greater health care resource utilization. Sickle cell anemia patients undergoing spinal fusion surgeries should be counseled on these increased risks. Moreover, current strategies for perioperative management of sickle cell anemia patients undergoing spinal fusion surgery need to be improved.
Collapse
|
31
|
Teixeira RS, Arriaga MB, Terse-Ramos R, Ferreira TA, Machado VR, Rissatto-Lago MR, Silveira-Mattos PS, Boa-Sorte N, Ladeia AMT, Andrade BB. Higher values of triglycerides:HDL-cholesterol ratio hallmark disease severity in children and adolescents with sickle cell anemia. ACTA ACUST UNITED AC 2019; 52:e8833. [PMID: 31618296 PMCID: PMC6799940 DOI: 10.1590/1414-431x20198833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/26/2019] [Indexed: 01/01/2023]
Abstract
Dyslipidemia has been described in sickle cell anemia (SCA) but its association with increased disease severity is unknown. Here, we examined 55 children and adolescents with SCA as well as 41 healthy controls to test the association between the lipid profiles in peripheral blood and markers of hemolysis, inflammation, endothelial function, and SCA-related clinical outcomes. SCA patients exhibited lower levels of total cholesterol (P<0.001), low-density lipoprotein cholesterol (LDL-c) (P<0.001), and high-density lipoprotein cholesterol (HDL-c) (P<0.001), while displaying higher triglyceride (TG) levels and TG/HDL-c ratio values (P<0.001). TG/HDL-c values were positively correlated with lactate dehydrogenase (P=0.047), leukocyte count (P=0.006), and blood flow velocity in the right (P=0.02) and left (P=0.05) cerebral artery, while being negatively correlated with hemoglobin levels (P<0.04). Acute chest syndrome (ACS) and vaso-occlusive events (VOE) were more frequent in SCA patients exhibiting higher TG/HDL-c values (odds ratio: 3.77, P=0.027). Multivariate logistic regression analysis confirmed independent associations between elevated TG/HDL-c values and SCA. Thus, children and adolescents with SCA exhibited a lipid profile associated with hemolysis and inflammatory parameters, with increased risk of ACS and VOE. TG/HDL-c is a potential biomarker of severity of disease.
Collapse
Affiliation(s)
- R S Teixeira
- Escola Bahiana de Medicina e Saúde Pública, Salvador, BA, Brasil.,Departamento de Pediatria, Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - M B Arriaga
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Fundação José Silveira, Salvador, BA, Brasil
| | - R Terse-Ramos
- Departamento de Pediatria, Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - T A Ferreira
- Departamento de Pediatria, Faculdade de Medicina, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - V R Machado
- Escola Bahiana de Medicina e Saúde Pública, Salvador, BA, Brasil
| | - M R Rissatto-Lago
- Departamento de Ciências da Vida, Universidade Estadual da Bahia, Salvador, BA, Brasil
| | - P S Silveira-Mattos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Fundação José Silveira, Salvador, BA, Brasil
| | - N Boa-Sorte
- Escola Bahiana de Medicina e Saúde Pública, Salvador, BA, Brasil.,Departamento de Ciências da Vida, Universidade Estadual da Bahia, Salvador, BA, Brasil
| | | | - B B Andrade
- Escola Bahiana de Medicina e Saúde Pública, Salvador, BA, Brasil.,Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brasil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Fundação José Silveira, Salvador, BA, Brasil.,Universidade Salvador, Laureate International Universities, Salvador, BA, Brasil
| |
Collapse
|
32
|
Ayoola OO, Bolarinwa RA, Onakpoya UU, Onwuka CC, Adedeji TA, Afolabi BI, Onigbinde SO, Arogundade FA. Doppler ultrasonographic evaluation of celiac and mesenteric arteries in subjects with sickle cell disease. JOURNAL OF CLINICAL ULTRASOUND : JCU 2019; 47:501-507. [PMID: 31063231 DOI: 10.1002/jcu.22729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/11/2019] [Accepted: 04/21/2019] [Indexed: 06/09/2023]
Abstract
Vasculopathy, as occurring in sickle cell disease (SCD), can affect celiac and mesenteric arteries and result in stenosis, with elevated peak systolic velocity (PSV) on Doppler ultrasonography. In six subjects with confirmed SCD in steady state, routine Doppler ultrasonographic examination discovered features of celiac artery (CA) or superior mesenteric artery (SMA) stenosis with CA PSV >200 cm/s (median = 222.8 cm/s; range = 201.5-427.1 cm/s) and/or SMA PSV >275 cm/s (median 183.2 cm/s; range = 87.8-289.3 cm/s). Among the six subjects, five had elevated soluble P-selectin values (median 72.55 ng/mL), while all six (100%) had elevated cystatin C levels (median 4.15 mg/L). Peripheral oxygen saturation was suboptimal in five subjects. All subjects had low hemoglobin concentration levels (median 8.5 g/dL) while four had elevated white blood cell count. Although vaso-occlusive crises result from microvessel occlusion, these findings at the macrovascular level suggest that SCD patients may also be vulnerable to mesenteric ischemic injury, especially in the setting of anemic heart failure from hemolysis.
Collapse
Affiliation(s)
- Oluwagbemiga O Ayoola
- Department of Radiology, Faculty of Clinical Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Rahman A Bolarinwa
- Department of Hematology and Blood Transfusion, Faculty of Basic Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Uvie U Onakpoya
- Department of Surgery, Faculty of Clinical Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Chidiogo C Onwuka
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Tewogbade A Adedeji
- Department of Chemical Pathology, Faculty of Basic Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Babalola I Afolabi
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Stephen O Onigbinde
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Fatiu A Arogundade
- Department of Internal Medicine, Faculty of Clinical Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| |
Collapse
|
33
|
Relating retinal blood flow and vessel morphology in sickle cell retinopathy. Eye (Lond) 2019; 34:886-891. [PMID: 31558825 PMCID: PMC7182580 DOI: 10.1038/s41433-019-0604-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/14/2019] [Indexed: 01/20/2023] Open
Abstract
Purpose The purpose of the current study was to determine associations between retinal blood flow and vessel morphology metrics in sickle cell retinopathy (SCR) and healthy normal control (NC) subjects. Methods Optical coherence tomography angiography (OCTA) and Doppler OCT imaging were performed in 12 SCR (15 eyes) and 19 NC (26 eyes) subjects. Vessel tortuosity was measured using a dedicated image analysis algorithm applied to OCTA images. Vessel density and spacing between vessels were determined from OCTA images by a fractal dimension analysis method. Retinal blood flow was quantified using a phase-resolved technique applied to en face Doppler OCT images. Results There was a significant association between increased retinal blood flow and increased vessel tortuosity (P = 0.03). Furthermore, increased retinal blood flow was associated with increased vessel density (P = 0.03) and decreased spacing between small vessels (P = 0.01). There was no significant association between retinal blood flow and spacing between large vessels (P = 0.11). Vessel tortuosity and blood flow were increased, whereas spacing between small vessels was decreased in SCR compared to NC group (P ≤ 0.03). There were no significant differences in vessel density or spacing between large vessels between the SCR and NC groups (P ≥ 0.31). Conclusions Associations between retinal hemodynamics and vessel morphology were reported, providing better understanding of retinal pathophysiology and insight into potential quantitative biomarkers to evaluate SCR.
Collapse
|
34
|
Mousavi Z, Yazdani Z, Moradabadi A, Hoseinpourkasgari F, Hassanshahi G. Role of some members of chemokine/cytokine network in the pathogenesis of thalassemia and sickle cell hemoglobinopathies: a mini review. Exp Hematol Oncol 2019; 8:21. [PMID: 31528501 PMCID: PMC6737600 DOI: 10.1186/s40164-019-0145-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/04/2019] [Indexed: 01/24/2023] Open
Abstract
The word of hemoglobinopathy is described for an array of disorders that affecting hemoglobin (Hb) functions. Hb is a molecule with 68 kDa molecular weight, serving as oxygen carrying metalloprotein. Hemoglobinopathy includes a wide range of Hb structural deficits varying from thalassemia to sickle cell disease. Cyto-chemokine network members are pivotally involved in the pathogenesis of hemoglobinopathies, however, the exact role of these mediators in the development of these disorders yet to be well addressed. Cytokines and chemokines are generated by inflamed endothelial cells that promote the expression of their respected receptors and further activate NF-κβ, recruit red blood cells (RBCs) and white blood cells (WBCs) toward the inflamed endothelium. Therefore, due to critical roles played by the cyto-chemokine network in several aspects of hemoglobinopathies pathophysiology including apoptosis of endothelial cells, RBC, WBC and etc.…, in the present review, we focused on the critical parts played by this network in the pathogenesis of hemoglobinopathies.
Collapse
Affiliation(s)
- Zahra Mousavi
- Department of Hematology and Medical Laboratory Sciences, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Zinat Yazdani
- Department of Hematology and Blood Banking, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Moradabadi
- Department of Hematology, School of Paramedicine, Arak University of Medical Science, Arak, Iran
| | | | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
35
|
Characterization of microvascular disease in patients with sickle cell disease using nailfold capillaroscopy. Microvasc Res 2019; 125:103877. [DOI: 10.1016/j.mvr.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 03/21/2019] [Accepted: 04/28/2019] [Indexed: 11/21/2022]
|
36
|
Juttukonda MR, Donahue MJ, Davis LT, Gindville MC, Lee CA, Patel NJ, Kassim AA, Pruthi S, Hendrikse J, Jordan LC. Preliminary evidence for cerebral capillary shunting in adults with sickle cell anemia. J Cereb Blood Flow Metab 2019; 39:1099-1110. [PMID: 29260615 PMCID: PMC6547194 DOI: 10.1177/0271678x17746808] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elevated flow velocities in adults with sickle cell anemia (SCA) may cause rapid erythrocyte transit through capillaries. This phenomenon could present as dural venous sinus hyperintensity on arterial spin labeling (ASL)-MRI and could be indicative of capillary shunting. Here, the prevalence of ASL venous hyperintensities and association with relevant physiology in adults with SCA was investigated. SCA ( n = 46) and age-matched control ( n = 16) volunteers were recruited for 3.0 T MRI. Pseudo-continuous ASL-MRI was acquired for cerebral blood flow (CBF) calculation and venous hyperintensity determination; venous signal intensity and a categorical venous score (three raters; 0 = no hyperintensity, 1 = focal hyperintensity, and 2 = diffuse hyperintensity) were recorded. Flow velocity in cervical internal carotid artery segments was determined from phase contrast data (venc = 40 cm/s) and whole-brain oxygen extraction fraction (OEF) was determined from T2-relaxation-under-spin-tagging MRI. Cerebral metabolic rate of oxygen was calculated as the product of OEF, CBF, and blood oxygen content. ASL venous hyperintensities were significantly ( p < 0.001) more prevalent in SCA (65%) relative to control (6%) participants and were associated with elevated flow velocities ( p = 0.03). CBF ( p < 0.001), but not OEF, increased with increasing hyperintensity score. Prospective trials that evaluate this construct as a possible marker of impaired oxygen delivery and stroke risk may be warranted.
Collapse
Affiliation(s)
- Meher R Juttukonda
- 1 Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manus J Donahue
- 1 Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,2 Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,3 Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA.,4 Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, USA
| | - Larry T Davis
- 1 Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melissa C Gindville
- 5 Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chelsea A Lee
- 5 Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Niral J Patel
- 5 Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adetola A Kassim
- 6 Department of Medicine, Division of Hematology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sumit Pruthi
- 1 Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeroen Hendrikse
- 7 Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lori C Jordan
- 2 Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,5 Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
37
|
McArthur JG, Svenstrup N, Chen C, Fricot A, Carvalho C, Nguyen J, Nguyen P, Parachikova A, Abdulla F, Vercellotti GM, Hermine O, Edwards D, Ribeil JA, Belcher JD, Maciel TT. A novel, highly potent and selective phosphodiesterase-9 inhibitor for the treatment of sickle cell disease. Haematologica 2019; 105:623-631. [PMID: 31147439 PMCID: PMC7049346 DOI: 10.3324/haematol.2018.213462] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
The most common treatment for patients with sickle cell disease (SCD) is the chemotherapeutic hydroxyurea, a therapy with pleiotropic effects, including increasing fetal hemoglobin (HbF) in red blood cells and reducing adhesion of white blood cells to the vascular endothelium. Hydroxyurea has been proposed to mediate these effects through a mechanism of increasing cellular cGMP levels. An alternative path to increasing cGMP levels in these cells is through the use of phosphodiesterase-9 inhibitors that selectively inhibit cGMP hydrolysis and increase cellular cGMP levels. We have developed a novel, potent and selective phosphodiesterase-9 inhibitor (IMR-687) specifically for the treatment of SCD. IMR-687 increased cGMP and HbF in erythroid K562 and UT-7 cells and increased the percentage of HbF positive erythroid cells generated in vitro using a two-phase liquid culture of CD34+ progenitors from sickle cell blood or bone marrow. Oral daily dosing of IMR-687 in the Townes transgenic mouse SCD model, increased HbF and reduced red blood cell sickling, immune cell activation and microvascular stasis. The IMR-687 reduction in red blood cell sickling and immune cell activation was greater than that seen with physiological doses of hydroxyurea. In contrast to other described phosphodiesterase-9 inhibitors, IMR-687 did not accumulate in the central nervous system, where it would inhibit phosphodiesterase-9 in neurons, or alter rodent behavior. IMR-687 was not genotoxic or myelotoxic and did not impact fertility or fetal development in rodents. These data suggest that IMR-687 may offer a safe and effective oral alternative for hydroxyurea in the treatment of SCD.
Collapse
Affiliation(s)
- James G McArthur
- Imara Inc., 2 Floor, 700 Technology Square, Cambridge, MA, USAImara Inc., 2
| | | | - Chunsheng Chen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Aurelie Fricot
- INSERM UMR 1163, CNRS ERL 8254, Imagine Institute, Laboratory of Excellence GR-Ex, Paris Descartes - Sorbonne Paris Cité University, Paris, France
| | - Caroline Carvalho
- INSERM UMR 1163, CNRS ERL 8254, Imagine Institute, Laboratory of Excellence GR-Ex, Paris Descartes - Sorbonne Paris Cité University, Paris, France
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Phong Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | | | - Fuad Abdulla
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Olivier Hermine
- INSERM UMR 1163, CNRS ERL 8254, Imagine Institute, Laboratory of Excellence GR-Ex, Paris Descartes - Sorbonne Paris Cité University, Paris, France
| | - Dave Edwards
- Kinexum, 8830 Glen Ferry Drive, Johns Creek, GA, USA
| | - Jean-Antoine Ribeil
- Departments of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris Descartes- Sorbonne Paris Cité University, Paris, France
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Thiago T Maciel
- INSERM UMR 1163, CNRS ERL 8254, Imagine Institute, Laboratory of Excellence GR-Ex, Paris Descartes - Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
38
|
Li X, Liu J, Hoh J, Liu J. Müller cells in pathological retinal angiogenesis. Transl Res 2019; 207:96-106. [PMID: 30639368 DOI: 10.1016/j.trsl.2018.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/06/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023]
Abstract
Müller cells are the major glial cells spanning the entire layer of the retina and maintaining retinal structure. Under pathological conditions, Müller cells are involved in retinal angiogenesis, a process of growing new blood vessels from pre-existing capillaries. In response to hypoxia, high glucose, and inflammation conditions, multiple signaling pathways are activated in Müller cells, followed by the increased production of proangiogenic factors including vascular endothelial growth factor, basic fibroblast growth factor, matrix metalloproteinases, Netrin-4, and angiopoietin-like 4. Expression of antiangiogenic factors is also downregulated in Müller cells. Besides, proliferation and dedifferentiation of Müller cells facilitates retinal angiogenesis. In this review, we summarized molecular mechanisms of Müller cells-related retinal angiogenesis. The potential of Müller cells as a therapeutic target for retinal angiogenesis was also discussed.
Collapse
Affiliation(s)
- Xiaorui Li
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China; Taishan Medical College, Taian, China
| | - Jing Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Josephine Hoh
- Department of Epidemiology and Public Health, Department of Ophthalmology and Visual Science, Yale University, New Haven, Connecticut
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
39
|
Uhelski ML, Simone DA. Sensitization of nociceptors and dorsal horn neurons contributes to pain in sickle cell disease. Neurosci Lett 2019; 705:20-26. [PMID: 30995520 DOI: 10.1016/j.neulet.2019.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/22/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Sickle cell disease (SCD) describes a group of disorders associated with a point mutation in the beta chain of hemoglobin. The mutation leads to the creation of sickle hemoglobin (HbS) and causes distortion of erythrocytes through polymerization under low oxygen, resulting in characteristic sickle red blood cells. Vaso-occlusion episodes caused by accumulation of sRBCs results in ischemia-reperfusion injury, reduced oxygen supply to organs, oxidative stress, organ damage and severe pain that often requires hospitalization and opioid treatment. Further, many patients suffer from chronic pain, including hypersensitivity to heat and cold stimuli. Progress towards the development of novel strategies for both acute and chronic pain in patients with SCD has been impeded by a lack of understanding the mechanisms underlying pain in SCD. The purpose of this review is to highlight evidence for the contribution of peripheral and central sensitization that leads to widespread, chronic pain and hyperalgesia. Targeting the mechanisms that initiate and maintain sensitization in SCD might offer effective approaches to manage the severe and debilitating pain associated with this condition.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Pain Medicine, Division of Anesthesiology, Critical Care and Pain Medicine. The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN 55455, United States.
| |
Collapse
|
40
|
Shah P, Khaleel M, Thuptimdang W, Sunwoo J, Veluswamy S, Chalacheva P, Kato RM, Detterich J, Wood JC, Zeltzer L, Sposto R, Khoo MCK, Coates TD. Mental stress causes vasoconstriction in subjects with sickle cell disease and in normal controls. Haematologica 2019; 105:83-90. [PMID: 30975906 PMCID: PMC6939522 DOI: 10.3324/haematol.2018.211391] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/05/2019] [Indexed: 01/02/2023] Open
Abstract
Vaso-occlusive crisis (VOC) is a hallmark of sickle cell disease (SCD) and occurs when deoxygenated sickled red blood cells occlude the microvasculature. Any stimulus, such as mental stress, which decreases microvascular blood flow will increase the likelihood of red cell entrapment resulting in local vaso-occlusion and progression to VOC. Neurally mediated vasoconstriction might be the physiological link between crisis triggers and vaso-occlusion. In this study, we determined the effect of mental stress on microvascular blood flow and autonomic nervous system reactivity. Sickle cell patients and controls performed mentally stressful tasks, including a memory task, conflict test and pain anticipation test. Blood flow was measured using photoplethysmography, autonomic reactivity was derived from electrocardiography and perceived stress was measured by the State-Trait Anxiety Inventory questionnaire. Stress tasks induced a significant decrease in microvascular blood flow, parasympathetic withdrawal and sympathetic activation in all subjects. Of the various tests, pain anticipation caused the highest degree of vasoconstriction. The magnitude of vasoconstriction, sympathetic activation and perceived stress was greater during the Stroop conflict test than during the N-back memory test, indicating the relationship between magnitude of experimental stress and degree of regional vasoconstriction. Baseline anxiety had a significant effect on the vasoconstrictive response in sickle cell subjects but not in controls. In conclusion, mental stress caused vasoconstriction and autonomic nervous system reactivity in all subjects. Although the pattern of responses was not significantly different between the two groups, the consequences of vasoconstriction can be quite significant in SCD because of the resultant entrapment of sickle cells in the microvasculature. This suggests that mental stress can precipitate a VOC in SCD by causing neural-mediated vasoconstriction.
Collapse
Affiliation(s)
- Payal Shah
- Division of Hematology, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - Maha Khaleel
- Division of Hematology, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - Wanwara Thuptimdang
- Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles
| | - John Sunwoo
- Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles
| | - Saranya Veluswamy
- Division of Hematology, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - Patjanaporn Chalacheva
- Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles
| | - Roberta M Kato
- Division of Pulmonology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - Jon Detterich
- Division of Cardiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - John C Wood
- Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles.,Division of Cardiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| | - Lonnie Zeltzer
- Pediatric Pain Program, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - Richard Sposto
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael C K Khoo
- Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles
| | - Thomas D Coates
- Division of Hematology, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
41
|
Gupta K, Chen C, Lutty GA, Hebbel RP. Morphine promotes neovascularizing retinopathy in sickle transgeneic mice. Blood Adv 2019; 3:1073-1083. [PMID: 30944099 PMCID: PMC6457224 DOI: 10.1182/bloodadvances.2018026898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Neovascularizing retinopathy is a significant complication of sickle cell disease (SCD), occurring more frequently in HbSC than HbSS disease. This risk difference is concordant with a divergence of angiogenesis risk, as identified by levels of pro- vs anti-angiogenic factors in the sickle patient's blood. Because our prior studies documented that morphine promotes angiogenesis in both malignancy and wound healing, we tested whether chronic opioid treatment would promote retinopathy in NY1DD sickle transgenic mice. After 10 to 15 months of treatment, sickle mice treated with morphine developed neovascularizing retinopathy to a far greater extent than either of the controls (sickle mice treated with saline and wild-type mice treated identically with morphine). Our dissection of the mechanistic linkage between morphine and retinopathy revealed a complex interplay among morphine engagement with its μ opioid receptor (MOR) on retinal endothelial cells (RECs); morphine-induced production of tumor necrosis factor α and interleukin-6 (IL-6), causing increased expression of both MOR and vascular endothelial growth factor receptor 2 (VEGFR2) on RECs; morphine/MOR engagement transactivating VEGFR2; and convergence of MOR, VEGFR2, and IL-6 activation on JAK/STAT3-dependent REC proliferation and angiogenesis. In the NY1DD mice, the result was increased angiogenesis, seen as neovascularizing retinopathy, similar to the retinal pathology occurring in humans with SCD. Therefore, we conclude that chronic opioid exposure, superimposed on the already angiogenic sickle milieu, might enhance risk for retinopathy. These results provide an additional reason for development and application of opioid alternatives for pain control in SCD.
Collapse
Affiliation(s)
- Kalpna Gupta
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| | - Chunsheng Chen
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| | - Gerard A Lutty
- Wilmer Ophthalmological Institute, John Hopkins School of Medicine, Baltimore, MD
| | - Robert P Hebbel
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| |
Collapse
|
42
|
Sensitization of nociceptors by prostaglandin E 2-glycerol contributes to hyperalgesia in mice with sickle cell disease. Blood 2019; 133:1989-1998. [PMID: 30796025 DOI: 10.1182/blood-2018-11-884346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/12/2019] [Indexed: 12/23/2022] Open
Abstract
Pain is a characteristic feature of sickle cell disease (SCD), 1 of the most common inherited diseases. Patients may experience acute painful crises as well as chronic pain. In the Berkley transgenic murine model of SCD, HbSS-BERK mice express only human hemoglobin S. These mice share many features of SCD patients, including persistent inflammation and hyperalgesia. Cyclooxygenase-2 (COX-2) is elevated in skin, dorsal root ganglia (DRG), and spinal cord in HbSS-BERK mice. In addition to arachidonic acid, COX-2 oxidizes the endocannabinoid 2-arachidonoylglycerol (2-AG) to produce prostaglandin E2 (PGE2)-glycerol (PGE2-G); PGE2-G is known to produce hyperalgesia. We tested the hypotheses that PGE2-G is increased in DRGs of HbSS-BERK mice and sensitizes nociceptors (sensory neurons that respond to noxious stimuli), and that blocking its synthesis would decrease hyperalgesia in HbSS-BERK mice. Systemic administration of R-flurbiprofen preferentially reduced production of PGE2-G over that of PGE2 in DRGs, decreased mechanical and thermal hyperalgesia, and decreased sensitization of nociceptors in HbSS-BERK mice. The same dose of R-flurbiprofen had no behavioral effect in HbAA-BERK mice (the transgenic control), but local injection of PGE2-G into the hind paw of HbAA-BERK mice produced sensitization of nociceptors and hyperalgesia. Coadministration of a P2Y6 receptor antagonist blocked the effect of PGE2-G, indicating that this receptor is a mediator of pain in SCD. The ability of R-flurbiprofen to block the synthesis of PGE2-G and to normalize levels of 2-AG suggests that R-flurbiprofen may be beneficial to treat pain in SCD, thereby reducing the use of opioids to relieve pain.
Collapse
|
43
|
Juttukonda MR, Donahue MJ. Neuroimaging of vascular reserve in patients with cerebrovascular diseases. Neuroimage 2019; 187:192-208. [PMID: 29031532 PMCID: PMC5897191 DOI: 10.1016/j.neuroimage.2017.10.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/01/2017] [Accepted: 10/07/2017] [Indexed: 12/21/2022] Open
Abstract
Cerebrovascular reactivity, defined broadly as the ability of brain parenchyma to adjust cerebral blood flow in response to altered metabolic demand or a vasoactive stimulus, is being measured with increasing frequency and may have a use for portending new or recurrent stroke risk in patients with cerebrovascular disease. The purpose of this review is to outline (i) the physiological basis of variations in cerebrovascular reactivity, (ii) available approaches for measuring cerebrovascular reactivity in research and clinical settings, and (iii) clinically-relevant cerebrovascular reactivity findings in the context of patients with cerebrovascular disease, including atherosclerotic arterial steno-occlusion, non-atherosclerotic arterial steno-occlusion, anemia, and aging. Literature references summarizing safety considerations for these procedures and future directions for standardizing protocols and post-processing procedures across centers are presented in the specific context of major unmet needs in the setting of cerebrovascular disease.
Collapse
Affiliation(s)
- Meher R Juttukonda
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
44
|
Ayoola OO, Bolarinwa RA, Onakpoya UU, Adedeji TA, Onwuka CC, Idowu BM. Intima-media thickness of the common femoral artery as a marker of leg ulceration in sickle cell disease patients. Blood Adv 2018; 2:3112-3117. [PMID: 30455360 PMCID: PMC6258917 DOI: 10.1182/bloodadvances.2018023267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
Leg ulceration is a debilitating chronic complication of sickle cell disease (SCD) the pathogenesis of which is yet to be fully elucidated. We hypothesized that SCD patients with histories of previous leg ulcers would have intima hyperplasia of the common femoral artery (CFA). We enrolled 44 SCD patients and 33 age-matched and sex-matched controls with hemoglobin AA. Anthropometric measurements, biochemical parameters, and sonographic intima-media thickness (IMT) of the CFA were determined. The median CFA IMT in SCD limbs with history of leg ulcers (SWLU) was 1.0 mm, whereas it was 0.7 mm in SCD limbs with no history of leg ulcer (SNLU) and 0.60 mm in controls (P < .001). Among the SNLU, 70.3% had CFA IMT <0.9 mm, whereas only 29.7% had CFA IMT ≥0.9 mm. Conversely, only 20.8% of SWLU had CFA IMT <0.9 mm, whereas the remaining 79.2% had CFA IMT ≥0.9 mm. All the controls had CFA IMT <0.9 mm. Binary logistic regression to determine the odds of having leg ulcer among SCD limbs with CFA IMT of ≥0.9 mm yielded an odds ratio of 9, indicating that SCD limbs with CFA IMT ≥0.9 mm had a 9 times greater risk of having leg ulcer compared with those with CFA IMT <0.9 mm. There is a significant increase in the CFA IMT of SCD limbs with ulcer compared with controls and SCD limbs without ulcer, suggesting that arterial vasculopathy plays a major role in the formation of these ulcers.
Collapse
Affiliation(s)
- Oluwagbemiga O Ayoola
- Department of Radiology, Faculty of Clinical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Osun State, Nigeria
| | | | | | - Tewogbade A Adedeji
- Deparment of Chemical Pathology, Faculty of Basic Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria; and
| | - Chidiogo C Onwuka
- Department of Radiology, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Osun State, Nigeria
| | - Bukunmi M Idowu
- Department of Radiology, Union Diagnostics and Clinical Services Plc, Yaba, Lagos, Nigeria
| |
Collapse
|
45
|
Juttukonda MR, Lee CA, Patel NJ, Davis LT, Waddle SL, Gindville MC, Pruthi S, Kassim AA, DeBaun MR, Donahue MJ, Jordan LC. Differential cerebral hemometabolic responses to blood transfusions in adults and children with sickle cell anemia. J Magn Reson Imaging 2018; 49:466-477. [PMID: 30324698 DOI: 10.1002/jmri.26213] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/18/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Blood transfusions are administered to children and adults with sickle cell anemia (SCA) for secondary stroke prevention, or as treatment for recurrent pain crises or acute anemia, but transfusion effects on cerebral hemodynamics and metabolism are not well-characterized. PURPOSE To compare blood transfusion-induced changes in hemometabolic parameters, including oxygen extraction fraction (OEF) and cerebral blood flow (CBF), within and between adults and children with SCA. STUDY TYPE Prospective, longitudinal study. SUBJECTS Adults with SCA (n = 16) receiving simple (n = 7) or exchange (n = 9) transfusions and children with SCA (n = 11) receiving exchange transfusions were scanned once when hematocrit was near nadir and again within 7 days of transfusion. Adult controls without SCA or sickle trait (n = 7) were scanned twice on separate days. FIELD STRENGTH/SEQUENCE 3.0T T1 -weighted, T2 -weighted, and T2 -relaxation-under-spin-tagging (TRUST) imaging, and phase contrast angiography. ASSESSMENT Global OEF was computed as the relative difference between venous oxygenation (from TRUST) and arterial oxygenation (from pulse oximetry). Global CBF was computed as total blood flow to the brain normalized by intracranial tissue volume. STATISTICAL TESTS Hemometabolic variables were compared using two-sided Wilcoxon signed-rank tests; associations were analyzed using two-sided Spearman's correlation testing. RESULTS In adults with SCA, posttransfusion OEF = 0.38 ± 0.05 was lower (P = 0.001) than pretransfusion OEF = 0.45 ± 0.09. A change in OEF was correlated with increases in hematocrit (P = 0.02; rho = -0.62) and with pretransfusion hematocrit (P = 0.02; rho = 0.65). OEF changes after transfusion were greater (P = 0.002) in adults receiving simple versus exchange transfusions. Posttransfusion CBF = 77.7 ± 26.4 ml/100g/min was not different (P = 0.27) from pretransfusion CBF = 82.3 ± 30.2 ml/100g/min. In children with SCA, both posttransfusion OEF = 0.28 ± 0.04 and CBF = 76.4 ± 26.4 were lower than pretransfusion OEF = 0.36 ± 0.06 (P = 0.004) and CBF = 96.4 ± 16.5 (P = 0.004). DATA CONCLUSION Cerebral OEF reduces following transfusions in adults and children with SCA. CBF reduces following transfusions more often in children compared to adults, indicating that vascular reserve capacity may remain near exhaustion posttransfusion in many adults. LEVEL OF EVIDENCE 2 Technical Efficacy Stage 5 J. Magn. Reson. Imaging 2019;49:466-477.
Collapse
Affiliation(s)
- Meher R Juttukonda
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Chelsea A Lee
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Niral J Patel
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry T Davis
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Spencer L Waddle
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Melissa C Gindville
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sumit Pruthi
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adetola A Kassim
- Department of Medicine, Division of Hematology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael R DeBaun
- Department of Pediatrics, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Psychiatry, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA
| | - Lori C Jordan
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Donahue MJ, Achten E, Cogswell PM, De Leeuw FE, Derdeyn CP, Dijkhuizen RM, Fan AP, Ghaznawi R, Heit JJ, Ikram MA, Jezzard P, Jordan LC, Jouvent E, Knutsson L, Leigh R, Liebeskind DS, Lin W, Okell TW, Qureshi AI, Stagg CJ, van Osch MJP, van Zijl PCM, Watchmaker JM, Wintermark M, Wu O, Zaharchuk G, Zhou J, Hendrikse J. Consensus statement on current and emerging methods for the diagnosis and evaluation of cerebrovascular disease. J Cereb Blood Flow Metab 2018; 38:1391-1417. [PMID: 28816594 PMCID: PMC6125970 DOI: 10.1177/0271678x17721830] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/26/2017] [Accepted: 06/10/2017] [Indexed: 01/04/2023]
Abstract
Cerebrovascular disease (CVD) remains a leading cause of death and the leading cause of adult disability in most developed countries. This work summarizes state-of-the-art, and possible future, diagnostic and evaluation approaches in multiple stages of CVD, including (i) visualization of sub-clinical disease processes, (ii) acute stroke theranostics, and (iii) characterization of post-stroke recovery mechanisms. Underlying pathophysiology as it relates to large vessel steno-occlusive disease and the impact of this macrovascular disease on tissue-level viability, hemodynamics (cerebral blood flow, cerebral blood volume, and mean transit time), and metabolism (cerebral metabolic rate of oxygen consumption and pH) are also discussed in the context of emerging neuroimaging protocols with sensitivity to these factors. The overall purpose is to highlight advancements in stroke care and diagnostics and to provide a general overview of emerging research topics that have potential for reducing morbidity in multiple areas of CVD.
Collapse
Affiliation(s)
- Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, USA
| | - Eric Achten
- Department of Radiology and Nuclear Medicine, Universiteit Gent, Gent, Belgium
| | - Petrice M Cogswell
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frank-Erik De Leeuw
- Radboud University, Nijmegen Medical Center, Donders Institute Brain Cognition & Behaviour, Center for Neuroscience, Department of Neurology, Nijmegen, The Netherlands
| | - Colin P Derdeyn
- Department of Radiology and Neurology, University of Iowa, Iowa City, IA, USA
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Audrey P Fan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Rashid Ghaznawi
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeremy J Heit
- Department of Radiology, Neuroimaging and Neurointervention Division, Stanford University, CA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Peter Jezzard
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Lori C Jordan
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric Jouvent
- Department of Neurology, AP-HP, Lariboisière Hospital, Paris, France
| | - Linda Knutsson
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Richard Leigh
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Weili Lin
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas W Okell
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Adnan I Qureshi
- Department of Neurology, Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Charlotte J Stagg
- Oxford Centre for Human Brain Activity, University of Oxford, Oxford, UK
| | | | - Peter CM van Zijl
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jennifer M Watchmaker
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Max Wintermark
- Department of Radiology, Neuroimaging and Neurointervention Division, Stanford University, CA, USA
| | - Ona Wu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Greg Zaharchuk
- Department of Radiology, Neuroimaging and Neurointervention Division, Stanford University, CA, USA
| | - Jinyuan Zhou
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
47
|
Chaturvedi S, Ghafuri DL, Jordan N, Kassim A, Rodeghier M, DeBaun MR. Clustering of end-organ disease and earlier mortality in adults with sickle cell disease: A retrospective-prospective cohort study. Am J Hematol 2018; 93:1153-1160. [PMID: 29981283 DOI: 10.1002/ajh.25202] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 01/10/2023]
Abstract
Chronic end-organ complications result in morbidity and mortality in adults with sickle cell disease (SCD). In a retrospective-prospective cohort of 150 adults with SCD who received standard care screening for pulmonary function abnormalities, cardiac disease, and renal assessment from January 2003 to 2016, we tested the hypothesis that clustering of end-organ disease is common and multiple organ impairment predicts mortality. Any end-organ disease occurred in 59.3% of individuals, and 24.0% developed multiple organ (>1) end-organ disease. The number of end-organs affected was associated with mortality (P ≤ .001); 8.2% (5 of 61) of individuals with no affected end-organ, 9.4% (5 of 53) of those with 1 affected organ, 20.7% (6 of 29) of those with 2 affected end-organs, and 85.7% (6 of 7) with 3 affected end-organs died over a median follow up period of 8.7 (interquartile range 3.5-11.4) years. Of the 22 individuals who died, 77.3% had evidence of any SCD-related end-organ impairment, and this was the primary or secondary cause of death in 45.0%. SCD-related chronic impairment in multiple organs, and its association with mortality, highlights the need to understand the common mechanisms underlying chronic end-organ damage in SCD, and the urgent need to develop interventions to prevent irreversible end-organ complications in SCD.
Collapse
Affiliation(s)
- Shruti Chaturvedi
- Division of Hematology, Department of Medicine; Johns Hopkins University School of Medicine; Nashville Tennesse
| | | | - Natalie Jordan
- Vanderbilt-Meharry Sickle Cell Disease Center of Excellence; Nashville Tennesse
- Division of Hematology and Oncology, Department of Medicine; Vanderbilt University Medical Center; Nashville Tennessee
| | - Adetola Kassim
- Vanderbilt-Meharry Sickle Cell Disease Center of Excellence; Nashville Tennesse
- Division of Hematology and Oncology, Department of Medicine; Vanderbilt University Medical Center; Nashville Tennessee
| | | | - Michael R. DeBaun
- Vanderbilt-Meharry Sickle Cell Disease Center of Excellence; Nashville Tennesse
- Department of Pediatrics; Vanderbilt University Medical Center; Nashville Tennesse
| |
Collapse
|
48
|
Musa BM, Odoh CN, Galadanci NA, Saidu H, Aliyu MH. Lower than expected elevated tricuspid regurgitant jet velocity in adults with sickle cell disease in Nigeria. Int Health 2018; 10:356-362. [PMID: 29438485 DOI: 10.1093/inthealth/ihx074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/13/2017] [Indexed: 11/15/2022] Open
Abstract
Background Cardiopulmonary disease is a major cause of morbidity and mortality in persons with sickle cell disease (SCD). Tricuspid regurgitant jet velocity (TRJV) and predicted forced expiratory volume in 1 s (FEV1%) predicted are independently associated with death in SCD. The goal of this study was to determine the prevalence of elevated TRJV and the association, if any, between TRJV and FEV1% predicted among persons with sickle cell anaemia (SCA) in Nigeria. Methods Using a cross-sectional design, we enrolled 100 adult Nigerians (≥15 y) with SCA. We screened participants using Doppler echocardiogram to determine their TRJV and assessed their lung function with spirometry. Results The prevalence of elevated TRJV was 6%, with 74% of participants having low FEV1% predicted (<70%). TRJV was negatively correlated with FEV1%, but this finding was not statistically significant (Spearman's ρ=-0.0263, p=0.8058). Conclusions We found a low prevalence of elevated TRJV and a trend in association between TRJV and FEV1% predicted in Nigerian adults with SCA. Our findings underscore the need to explore further the relationship between SCD and cardiopulmonary disease in adults.
Collapse
Affiliation(s)
- Baba Maiyaki Musa
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Chisom N Odoh
- Department of Epidemiology and Population Health, University of Louisville, Louisville, Kentucky, USA
| | - Najibah A Galadanci
- Department of Hematology, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Hadiza Saidu
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Muktar H Aliyu
- Vanderbilt Institute for Global Health, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Health Policy, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
49
|
Ribeil J, Labopin M, Stanislas A, Deloison B, Lemercier D, Habibi A, Albinni S, Charlier C, Lortholary O, Lefrere F, De Montalembert M, Blanche S, Galactéros F, Tréluyer J, Gluckman E, Ville Y, Joseph L, Delville M, Benachi A, Cavazzana M. Transfusion-related adverse events are decreased in pregnant women with sickle cell disease by a change in policy from systematic transfusion to prophylactic oxygen therapy at home: A retrospective survey by the international sickle cell disease observatory. Am J Hematol 2018; 93:794-802. [PMID: 29603363 PMCID: PMC6001537 DOI: 10.1002/ajh.25097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 02/02/2023]
Abstract
Sickle cell disease (SCD) in pregnancy can be associated with adverse maternal and perinatal outcomes. Furthermore, complications of SCD can be aggravated by pregnancy. Optimal prenatal care aims to decrease the occurrence of maternal and fetal complications. A retrospective, French, two-center study compared two care strategies for pregnant women with SCD over two time periods. In the first study period (2005-2010), the women were systematically offered prophylactic transfusions. In the second study period (2011-2014), a targeted transfusion strategy was applied whenever possible, and home-based prophylactic nocturnal oxygen therapy was offered to all the pregnant women. The two periods did not differ significantly in terms of the incidence of vaso-occlusive events. Maternal mortality, perinatal mortality, and obstetric complication rates were also similar in the two periods, as was the incidence of post-transfusion complications (6.1% in 2005-2010 and 1.3% in 2011-2014, P = .15), although no de novo alloimmunizations or delayed hemolysis transfusion reactions were observed in the second period. The results of this preliminary, retrospective study indicate that targeted transfusion plus home-based prophylactic nocturnal oxygen therapy is safe and may decrease transfusion requirements and transfusion-associated complications.
Collapse
Affiliation(s)
- Jean‐Antoine Ribeil
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- Biotherapy CIC, West University Hospital Group, Assistance Publique‐Hôpitaux de Paris, INSERMParisFrance
| | - Myriam Labopin
- Clinical Hematology and Cellular Therapy DepartmentSaint‐Antoine Hospital, Assistance Publique‐Hôpitaux de Paris, France ‐ INSERM UMRs 938, Pierre et Marie Curie University (UPMC, Paris VI)ParisFrance
| | - Aurélie Stanislas
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- Biotherapy CIC, West University Hospital Group, Assistance Publique‐Hôpitaux de Paris, INSERMParisFrance
| | - Benjamin Deloison
- Department of Obstetrics and Fetal MedicineNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Delphine Lemercier
- Department of Obstetrics and Fetal MedicineNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Anoosha Habibi
- Reference Center for Sickle Cell Disease, Henri Mondor Hospital, Assistance Publique‐Hôpitaux de ParisCréteilFrance
| | - Souha Albinni
- Necker Children's HospitalFrench Blood Establishment ‐ Ile de FranceParisFrance
| | - Caroline Charlier
- Necker Children's Hospital, Assistance Publique‐Hôpitaux de ParisNecker Pasteur Center for Infectious Diseases and Tropical MedicineParisFrance
| | - Olivier Lortholary
- Imagine InstituteParisFrance
- Necker Children's Hospital, Assistance Publique‐Hôpitaux de ParisNecker Pasteur Center for Infectious Diseases and Tropical MedicineParisFrance
- Paris Descartes UniversityParisFrance
| | - François Lefrere
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- Biotherapy CIC, West University Hospital Group, Assistance Publique‐Hôpitaux de Paris, INSERMParisFrance
| | - Mariane De Montalembert
- Reference Centre for Sickle Cell Disease, Pediatric DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Stéphane Blanche
- Unit of Pediatric Immunology and HematologyNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Frédéric Galactéros
- Reference Center for Sickle Cell Disease, Henri Mondor Hospital, Assistance Publique‐Hôpitaux de ParisCréteilFrance
| | - Jean‐Marc Tréluyer
- Paris Descartes UniversityParisFrance
- Necker Children's Hospital, Assistance Publique‐Hôpitaux de ParisClinical Research Unit/Clinical Investigation CentreParisFrance
| | - Eliane Gluckman
- Saint‐Louis Hospital, Paris, France and Monaco Scientific CenterEurocord Monacord International Observatory on Sickle Cell DiseaseMonaco
| | - Yves Ville
- Department of Obstetrics and Fetal MedicineNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Laure Joseph
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Marianne Delville
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| | - Alexandra Benachi
- Department of Obstetrics and Gynecology and Reproductive MedicineAntoine Béclère Hospital, Assistance Publique‐Hôpitaux de Paris, Université Paris SudClamartFrance
| | - Marina Cavazzana
- Biotherapy DepartmentNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- Biotherapy CIC, West University Hospital Group, Assistance Publique‐Hôpitaux de Paris, INSERMParisFrance
- Paris Descartes UniversityParisFrance
- Unit of Pediatric Immunology and HematologyNecker Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- Saint‐Louis Hospital, Paris, France and Monaco Scientific CenterEurocord Monacord International Observatory on Sickle Cell DiseaseMonaco
| |
Collapse
|
50
|
Sensitization of C-fiber nociceptors in mice with sickle cell disease is decreased by local inhibition of anandamide hydrolysis. Pain 2018; 158:1711-1722. [PMID: 28570479 DOI: 10.1097/j.pain.0000000000000966] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Chronic pain and hyperalgesia, as well as pain resulting from episodes of vaso-occlusion, are characteristic features of sickle cell disease (SCD) and are difficult to treat. Since there is growing evidence that increasing local levels of endocannabinoids can decrease hyperalgesia, we examined the effects of URB597, a fatty acid amide hydrolase (FAAH) inhibitor, which blocks the hydrolysis of the endogenous cannabinoid anandamide, on hyperalgesia and sensitization of cutaneous nociceptors in a humanized mouse model of SCD. Using homozygous HbSS-BERK sickle mice, we determined the effects of URB597 on mechanical hyperalgesia and on sensitization of C-fiber nociceptors in vivo. Intraplantar administration of URB597 (10 μg in 10 μL) decreased the frequency of withdrawal responses evoked by a von Frey monofilament (3.9 mN bending force) applied to the plantar hind paw. This was blocked by the CB1 receptor antagonist AM281 but not by the CB2 receptor antagonist AM630. Also, URB597 decreased hyperalgesia in HbSS-BERK/CB2R sickle mice, further confirming the role of CB1 receptors in the effects produced by URB597. Electrophysiological recordings were made from primary afferent fibers of the tibial nerve in anesthetized mice. The proportion of Aδ- and C-fiber nociceptors that exhibited spontaneous activity and responses of C-fibers to mechanical and thermal stimuli were greater in HbSS-BERK sickle mice as compared to control HbAA-BERK mice. Spontaneous activity and evoked responses of nociceptors were decreased by URB597 via CB1 receptors. It is suggested that enhanced endocannabinoid activity in the periphery may be beneficial in alleviating chronic pain associated with SCD.
Collapse
|