1
|
Nakagawa A, Morwood K, Johnson K, Berra L, Boal L, Azar S, Huang M, Heeney MM, Bloch DB, Ichinose F. Screen of the ReFRAME Compound Library for Therapeutic Agents to Prevent Red Blood Cell Sickling Using an Improved High Throughput Sickling Assay. ACS OMEGA 2025; 10:16497-16505. [PMID: 40321528 PMCID: PMC12044578 DOI: 10.1021/acsomega.4c11077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/30/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
Sickle cell disease (SCD) is an autosomal recessive disorder of blood characterized by a mutation in the β chain of hemoglobin (Hb), leading to the production of sickle Hb (HbS). In SCD, under low oxygen conditions, red blood cells (RBCs) containing HbS form a characteristic "sickle" shape, resulting in chronic hemolytic anemia and acute vaso-occlusive crises. Current therapies for SCD have limitations in efficacy or availability, highlighting the need for new anti-sickling drugs. To facilitate the discovery of new anti-sickling compounds, we previously developed a high throughput sickling assay, which permits rapid screening of thousands of compounds for the ability to inhibit RBC sickling. In this study, we improved the sickling assay by optimizing the assay condition and expanded our screening efforts by evaluating the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) compound library, which contains approximately 2.5 times more compounds than previously screened. We were able to increase the number of blood samples that were adequate for identifying anti-sickling compounds in the improved sickling assay and identified voxelotor and SNS-314 as compounds that successfully prevented sickling. The improved sickling assay will increase access to valuable blood samples from SCD volunteers, providing more opportunities to develop anti-sickling compounds for treating SCD.
Collapse
Affiliation(s)
- Akito Nakagawa
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Kaycie Morwood
- The
Calibr-Skaggs Institute for Innovative Medicines, Scripps Research, San Diego, California 92037, United States
| | - Kristen Johnson
- The
Calibr-Skaggs Institute for Innovative Medicines, Scripps Research, San Diego, California 92037, United States
| | - Lorenzo Berra
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lauren Boal
- Division
of Pediatric Hematology and Oncology, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Sharl Azar
- Comprehensive
Sickle Cell Disease Treatment Center, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mary Huang
- Division
of Pediatric Hematology and Oncology, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Matthew M. Heeney
- Dana-Farber/Boston
Children’s Cancer and Blood Disorders Center and Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Donald B. Bloch
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Division
of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Fumito Ichinose
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
2
|
Galamba N. Membrane Permeability of Cyclic and Linear Peptides, a Halogenated Antisickling Molecule, and Water Across a Red Blood Cell Bilayer Model. J Phys Chem Lett 2025; 16:4021-4030. [PMID: 40228479 DOI: 10.1021/acs.jpclett.4c03339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
We studied through molecular dynamics and inhomogeneous solubility-diffusion theory the permeability of several cyclic peptides (CPs) recently proposed as potential antisickling drugs, across a red blood cell (RBC) membrane model. The permeability of the CPs is compared to that of a linear precursor, a highly charged CP, a high permeability halogenated antisickling molecule (PD150606), and water. The influence of cholesterol (45% of the membrane) is assessed through comparison with the permeability across a homogeneous lipid bilayer. The most promising CPs concerning their potential antisickling activity depict the highest permeabilities, only exceeded by PD150606. The permeability of a hydrophobic CP is four decades higher than its linear precursor despite noncovalent cyclization in the interior of the membrane. Further, cholesterol is found to significantly reduce the permeability of water and a model CP, while not influencing that of PD150606. The influence of the water model is also investigated.
Collapse
Affiliation(s)
- N Galamba
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
3
|
Goel Y, Arellano MA, Fouda RT, Garcia NR, Lomeli RA, Kerr D, Argueta DA, Gupta M, Velasco GJ, Prince R, Banerjee P, Jana S, Alayash AI, Friedman JM, Gupta K. Targeting sickle cell pathobiology and pain with novel transdermal curcumin. PNAS NEXUS 2025; 4:pgaf053. [PMID: 40007577 PMCID: PMC11854080 DOI: 10.1093/pnasnexus/pgaf053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025]
Abstract
Several comorbidities of sickle cell disease (SCD) originate from red blood cell (RBC) instability, chronic inflammation, and oxidative stress. Development of scalable, cost-effective therapeutics suitable for chronic administration to prevent, attenuate, and perhaps reverse the consequences of RBC instability is needed. Curcumin has many of these attributes as a safe compound with antisickling, antiinflammatory, and antioxidant properties, but its translational potential has been constrained due to limited bioavailability from oral administration. The present study demonstrates the rapid and high bioavailability of a novel topical/transdermal (TD) curcumin gel formulation in the plasma and blood cells and its effectiveness in humanized sickle cell mice in: (i) ameliorating features of sickle cell pain hypersensitivity and axonal injury; (ii) reducing multiple manifestations of RBC instability including evidence of decreased hemolysis (reduced lactate dehydrogenase levels), enhanced RBC ATP levels along with decreased oxidative damage; (iii) decreasing multiple proinflammatory cytokines including interleukin-6, monocyte chemoattractant protein-1, granulocyte-macrophage colony-stimulating factor, and activation, normal T cell expressed and secreted protein in skin secretome; and (iv) reducing mast cell degranulation and activation. Our data suggest that an easy-to-use novel TD curcumin gel formulation has the potential to ameliorate chronic pain, improve RBC stability, and reduce inflammatory consequences of SCD.
Collapse
Affiliation(s)
- Yugal Goel
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Mya A Arellano
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Raghda T Fouda
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Natalie R Garcia
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Reina A Lomeli
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Daniel Kerr
- Department of Chemistry and Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314, USA
| | - Donovan A Argueta
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Mihir Gupta
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Graham J Velasco
- Pathology Department, VA Long Beach Medical Center, Long Beach, CA 90822, USA
| | | | - Probal Banerjee
- Department of Chemistry and Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314, USA
| | - Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Joel M Friedman
- Vascarta, Inc., Summit, NJ 07446, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kalpna Gupta
- Hematology/Oncology Division, Department of Medicine, University of California, Irvine, CA 92697, USA
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
4
|
Alaimo B, Metaferia B, Cellmer T, Thein SL, Bunn HF, Eaton WA. Therapeutic potential of the latest oxygen affinity-modifying agent, GBT021601, for treating sickle cell disease is questionable. Br J Haematol 2024; 205:2527-2529. [PMID: 39503354 PMCID: PMC11637734 DOI: 10.1111/bjh.19697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 11/08/2024]
Affiliation(s)
- Braydon Alaimo
- Laboratory of Chemical PhysicsNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Belhu Metaferia
- Laboratory of Chemical PhysicsNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Troy Cellmer
- Laboratory of Chemical PhysicsNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Swee Lay Thein
- Sickle Cell BranchNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - H. Franklin Bunn
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - William A. Eaton
- Laboratory of Chemical PhysicsNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
5
|
Galamba N. Sickle Cell Hemoglobin "Drugged" with Cyclic Peptides Is Aggregation Incompetent. J Phys Chem B 2024; 128:8662-8671. [PMID: 39205400 PMCID: PMC11403655 DOI: 10.1021/acs.jpcb.4c03805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Sickle cell disease (SCD) is a monogenic blood disorder associated with a mutation in the hemoglobin subunit β gene encoding for the β-globin of normal adult hemoglobin (HbA). This mutation transcribes into a Glu-β6 → Val-β6 substitution in the β-globins, inducing the polymerization of this hemoglobin form (HbS) when in the T-state. Despite advances in stem cell and gene therapy, and the recent approval of a new antisickling drug, therapeutic limitations persist. Herein, we demonstrate through molecular dynamics and umbrella sampling, that (unrestrained) blockage of the hydrophobic pocket involved in the lateral contact of the HbS fibers by 5-mer cyclic peptides, recently proposed as SCD aggregation inhibitors (Neto, V.; J. Med. Chem. 2023, 66, 16062-16074), is enough to turn the dimerization of HbS thermodynamically unfavorable. Among these potential drugs, some exhibit an estimated pocket abandonment probability of around 15-20% within the simulations' time frame, and an impressive specificity toward the mutated Val-β6. Additionally, we show that the dimerization can be thermodynamically unfavored by blocking a nearby region while the pocket remains vacant. These results are compared with curcumin, an antisickling molecule and a pan-assay interference compound, with a good binding affinity for different proteins and protein domains. Our results confirm the potential of some of these cyclic peptides as antisickling drug candidates to reduce the concentration of aggregation-competent HbS.
Collapse
Affiliation(s)
- N. Galamba
- Biosystems and Integrative
Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
6
|
D'Alessandro A, Le K, Lundt M, Li Q, Dunkelberger EB, Cellmer T, Worth AJ, Patil S, Huston C, Grier A, Dzieciatkowska M, Stephenson D, Eaton WA, Thein SL. Functional and multi-omics signatures of mitapivat efficacy upon activation of pyruvate kinase in red blood cells from patients with sickle cell disease. Haematologica 2024; 109:2639-2652. [PMID: 38450513 PMCID: PMC11290518 DOI: 10.3324/haematol.2023.284831] [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: 12/11/2023] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Mitapivat, a pyruvate kinase activator, shows great potential as a sickle cell disease (SCD)-modifying therapy. The safety and efficacy of mitapivat as a long-term maintenance therapy are currently being evaluated in two open-label studies. Here we applied a comprehensive multi-omics approach to investigate the impact of activating pyruvate kinase on red blood cells (RBC) from 15 SCD patients. HbSS patients were enrolled in one of the open-label, extended studies (NCT04610866). Leukodepleted RBC obtained from fresh whole blood at baseline, prior to drug initiation, and at longitudinal timepoints over the course of the study were processed for multi-omics through a stepwise extraction of metabolites, lipids and proteins. Mitapivat therapy had significant effects on the metabolome, lipidome and proteome of SCD RBC. Mitapivat decreased 2,3-diphosphoglycerate levels, increased adenosine triphosphate levels, and improved hematologic and sickling parameters in patients with SCD. Agreement between omics measurements and clinical measurements confirmed the specificity of mitapivat on targeting late glycolysis, with glycolytic metabolites ranking as the top correlates to parameters of hemoglobin S oxygen affinity (p50) and sickling kinetics (t50) during treatment. Mitapivat markedly reduced levels of proteins of mitochondrial origin within 2 weeks of initiation of treatment, with minimal changes in reticulocyte counts. In the first 6 months of treatment there were also transient elevations of lysophosphatidylcholines and oxylipins with depletion of free fatty acids, suggestive of an effect on membrane lipid remodeling. Multi-omics analysis of RBC identified benefits for glycolysis, as well as activation of the Lands cycle.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO.
| | - Kang Le
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda
| | - Maureen Lundt
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda
| | - Quan Li
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda
| | - Emily B Dunkelberger
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda
| | - Troy Cellmer
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda
| | | | | | | | - Abby Grier
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO
| | - William A Eaton
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda.
| |
Collapse
|
7
|
Leonard AK, Furstenau D, Inam Z, Luckett C, Chu R, Demirci S, Essawi K, Gudmundsdottir B, Hinds M, DiNicola J, Li Q, Eaton WA, Cellmer T, Wang X, Thein SL, Macari ER, VanNest S, Hsieh MM, Bonner M, Pierciey FJ, Tisdale JF. In vivo measurement of RBC survival in patients with sickle cell disease before or after hematopoietic stem cell transplantation. Blood Adv 2024; 8:1806-1816. [PMID: 38181784 PMCID: PMC11006808 DOI: 10.1182/bloodadvances.2023011397] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/17/2023] [Accepted: 11/11/2023] [Indexed: 01/07/2024] Open
Abstract
ABSTRACT Stable, mixed-donor-recipient chimerism after allogeneic hematopoietic stem cell transplantation (HSCT) for patients with sickle cell disease (SCD) is sufficient for phenotypic disease reversal, and results from differences in donor/recipient-red blood cell (RBC) survival. Understanding variability and predictors of RBC survival among patients with SCD before and after HSCT is critical for gene therapy research which seeks to generate sufficient corrected hemoglobin to reduce polymerization thereby overcoming the red cell pathology of SCD. This study used biotin labeling of RBCs to determine the lifespan of RBCs in patients with SCD compared with patients who have successfully undergone curative HSCT, participants with sickle cell trait (HbAS), and healthy (HbAA) donors. Twenty participants were included in the analysis (SCD pre-HSCT: N = 6, SCD post-HSCT: N = 5, HbAS: N = 6, and HbAA: N = 3). The average RBC lifespan was significantly shorter for participants with SCD pre-HSCT (64.1 days; range, 35-91) compared with those with SCD post-HSCT (113.4 days; range, 105-119), HbAS (126.0 days; range, 119-147), and HbAA (123.7 days; range, 91-147) (P<.001). RBC lifespan correlated with various hematologic parameters and strongly correlated with the average final fraction of sickled RBCs after deoxygenation (P<.001). No adverse events were attributable to the use of biotin and related procedures. Biotin labeling of RBCs is a safe and feasible methodology to evaluate RBC survival in patients with SCD before and after HSCT. Understanding differences in RBC survival may ultimately guide gene therapy protocols to determine hemoglobin composition required to reverse the SCD phenotype as it relates directly to RBC survival. This trial was registered at www.clinicaltrials.gov as #NCT04476277.
Collapse
Affiliation(s)
- Alexis K. Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Dana Furstenau
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
- Department of Pediatrics, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zaina Inam
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
| | - Christina Luckett
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Rebecca Chu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Bjorg Gudmundsdottir
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Malikiya Hinds
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Julia DiNicola
- Cellular and Molecular Therapeutics 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, Bethesda, MD
| | - William A. Eaton
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Troy Cellmer
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Xunde Wang
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Matthew M. Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
8
|
Qiang Y, Xu M, Patel Pochron M, Jupelli M, Dao M. A framework of computer vision-enhanced microfluidic approach for automated assessment of the transient sickling kinetics in sickle red blood cells. FRONTIERS IN PHYSICS 2024; 12:1331047. [PMID: 38605818 PMCID: PMC11008125 DOI: 10.3389/fphy.2024.1331047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The occurrence of vaso-occlusive crisis greatly depends on the competition between the sickling delay time and the transit time of individual sickle cells, i.e., red blood cells (RBCs) from sickle cell disease (SCD) patients, while they are traversing the circulatory system. Many drugs for treating SCD work by inhibiting the polymerization of sickle hemoglobin (HbS), effectively delaying the sickling process in sickle cells (SS RBCs). Most previous studies on screening anti-sickling drugs, such as voxelotor, rely on in vitro testing of sickling characteristics, often conducted under prolonged deoxygenation for up to 1 hour. However, since the microcirculation of RBCs typically takes less than 1 minute, the results of these studies may be less accurate and less relevant for in vitro-in vivo correlation. In our current study, we introduce a computer vision-enhanced microfluidic framework designed to automatically capture the transient sickling kinetics of SS RBCs within a 1-min timeframe. Our study has successfully detected differences in the transient sickling kinetics between vehicle control and voxelotor-treated SS RBCs. This approach has the potential for broader applications in screening anti-sickling therapies.
Collapse
Affiliation(s)
- Yuhao Qiang
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Mengjia Xu
- Department of Data Science, Ying Wu College of Computing, New Jersey Institute of Technology, Newark, NJ, United States
- Center for Brains, Minds and Machines, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | | | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
9
|
Parekh DS, Eaton WA, Thein SL. Recent developments in the use of pyruvate kinase activators as a new approach for treating sickle cell disease. Blood 2024; 143:866-871. [PMID: 38118071 PMCID: PMC10940061 DOI: 10.1182/blood.2023021167] [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: 09/11/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 12/22/2023] Open
Abstract
ABSTRACT Pyruvate kinase (PK) is a key enzyme in glycolysis, the sole source of adenosine triphosphate, which is essential for all energy-dependent activities of red blood cells. Activating PK shows great potential for treating a broad range of hemolytic anemias beyond PK deficiency, because they also enhance activity of wild-type PK. Motivated by observations of sickle-cell complications in sickle-trait individuals with concomitant PK deficiency, activating endogenous PK offers a novel and promising approach for treating patients with sickle-cell disease.
Collapse
Affiliation(s)
- Dina S. Parekh
- Sickle Cell Branch, 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
|
10
|
Gupta P, Goswami SG, Kumari G, Saravanakumar V, Bhargava N, Rai AB, Singh P, Bhoyar RC, Arvinden VR, Gunda P, Jain S, Narayana VK, Deolankar SC, Prasad TSK, Natarajan VT, Scaria V, Singh S, Ramalingam S. Development of pathophysiologically relevant models of sickle cell disease and β-thalassemia for therapeutic studies. Nat Commun 2024; 15:1794. [PMID: 38413594 PMCID: PMC10899644 DOI: 10.1038/s41467-024-46036-x] [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: 03/12/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
Ex vivo cellular system that accurately replicates sickle cell disease and β-thalassemia characteristics is a highly sought-after goal in the field of erythroid biology. In this study, we present the generation of erythroid progenitor lines with sickle cell disease and β-thalassemia mutation using CRISPR/Cas9. The disease cellular models exhibit similar differentiation profiles, globin expression and proteome dynamics as patient-derived hematopoietic stem/progenitor cells. Additionally, these cellular models recapitulate pathological conditions associated with both the diseases. Hydroxyurea and pomalidomide treatment enhanced fetal hemoglobin levels. Notably, we introduce a therapeutic strategy for the above diseases by recapitulating the HPFH3 genotype, which reactivates fetal hemoglobin levels and rescues the disease phenotypes, thus making these lines a valuable platform for studying and developing new therapeutic strategies. Altogether, we demonstrate our disease cellular systems are physiologically relevant and could prove to be indispensable tools for disease modeling, drug screenings and cell and gene therapy-based applications.
Collapse
Affiliation(s)
- Pragya Gupta
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sangam Giri Goswami
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Geeta Kumari
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Vinodh Saravanakumar
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
| | - Nupur Bhargava
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
| | - Akhila Balakrishna Rai
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Praveen Singh
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rahul C Bhoyar
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
| | - V R Arvinden
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Padma Gunda
- Thalassemia and Sickle Cell Society- Kamala Hospital and Research Centre, Shivarampally, Hyderabad, India
| | - Suman Jain
- Thalassemia and Sickle Cell Society- Kamala Hospital and Research Centre, Shivarampally, Hyderabad, India
| | - Vanya Kadla Narayana
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Sayali C Deolankar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Vivek T Natarajan
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vinod Scaria
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shailja Singh
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| | - Sivaprakash Ramalingam
- CSIR- Institute for Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Neto V, Victor BL, Galamba N. Cyclic Peptides as Aggregation Inhibitors for Sickle Cell Disease. J Med Chem 2023; 66:16062-16074. [PMID: 37988411 DOI: 10.1021/acs.jmedchem.3c01484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Sickle cell disease is a missense genetic disorder characterized by the aggregation of deoxy-HbS into helical fibers that distort erythrocytes into a sickle-like shape. Herein, we investigate, through molecular dynamics, the effect of nine 5-mer cyclic peptides (CPs), tailor-designed to block key lateral contacts of the fibers. Our results show that the CPs bind orthogonally to the main HbS pocket involved in the latter contacts, with some revealing exceedingly long residence times. These CPs display moderate to high specificity, exhibiting molecular recognition events even at a HbS/CP (1:1) ratio. A much lower HbS-CP binding free energy, longer residence times, and higher specificity are also found relative to a previously reported CP with modest in vitro antisickling activity. These results indicate that some of these CPs have the potential to reduce the concentration of aggregation-competent deoxy-HbS, precluding or delaying the formation of lateral contact at the homogeneous nucleation stage.
Collapse
Affiliation(s)
- Vasco Neto
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande 1749-016, Lisboa, Portugal
| | - Bruno Lourenço Victor
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande 1749-016, Lisboa, Portugal
| | - Nuno Galamba
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Edifício C8, Campo Grande 1749-016, Lisboa, Portugal
| |
Collapse
|
12
|
Kawata M, McClatchy DB, Diedrich JK, Olmer M, Johnson KA, Yates JR, Lotz MK. Mocetinostat activates Krüppel-like factor 4 and protects against tissue destruction and inflammation in osteoarthritis. JCI Insight 2023; 8:e170513. [PMID: 37681413 PMCID: PMC10544226 DOI: 10.1172/jci.insight.170513] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder, and disease-modifying OA drugs (DMOADs) represent a major need in OA management. Krüppel-like factor 4 (KLF4) is a central transcription factor upregulating regenerative and protective functions in joint tissues. This study was aimed to identify small molecules activating KLF4 expression and to determine functions and mechanisms of the hit compounds. High-throughput screening (HTS) with 11,948 clinical-stage compounds was performed using a reporter cell line detecting endogenous KLF4 activation. Eighteen compounds were identified through the HTS and confirmed in a secondary screen. After testing in SW1353 chondrosarcoma cells and human chondrocytes, mocetinostat - a class I selective histone deacetylase (HDAC) inhibitor - had the best profile of biological activities. Mocetinostat upregulated cartilage signature genes in human chondrocytes, meniscal cells, and BM-derived mesenchymal stem cells, and it downregulated hypertrophic, inflammatory, and catabolic genes in those cells and synoviocytes. I.p. administration of mocetinostat into mice reduced severity of OA-associated changes and improved pain behaviors. Global gene expression and proteomics analyses revealed that regenerative and protective effects of mocetinostat were dependent on peroxisome proliferator-activated receptor γ coactivator 1-α. These findings show therapeutic and protective activities of mocetinostat against OA, qualifying it as a candidate to be used as a DMOAD.
Collapse
Affiliation(s)
- Manabu Kawata
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Daniel B. McClatchy
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Jolene K. Diedrich
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | | | - John R. Yates
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Martin K. Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
13
|
Worth EH, Fugate MK, Ferrone FA. Voxelotor does not inhibit sickle hemoglobin fiber formation upon complete deoxygenation. Biophys J 2023; 122:2782-2790. [PMID: 37270670 PMCID: PMC10397806 DOI: 10.1016/j.bpj.2023.05.034] [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: 02/01/2023] [Revised: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
The drug voxelotor (commercially known as Oxbryta) has been approved by the US Food and Drug Administration for the treatment of sickle cell disease. It is known to reduce disease-causing sickling by inhibiting the transformation of the non-polymerizing, high-oxygen-affinity R quaternary structure of sickle hemoglobin into its polymerizing, low-affinity T quaternary structure. It has not been established whether the binding of the drug has anti-sickling effects beyond restricting the change of quaternary structure. By using a laser photolysis method that employs microscope optics, we have determined that fully deoxygenated sickle hemoglobin will assume the T structure. We show that the nucleation rates essential to generate the sickle fibers are not significantly affected by voxelotor. The method employed here should be useful for determining the mechanism of sickling inhibition for proposed drugs.
Collapse
Affiliation(s)
- Eli H Worth
- Department of Physics, Drexel University, Philadelphia, Pennsylvania
| | - Mark K Fugate
- Department of Physics, Drexel University, Philadelphia, Pennsylvania
| | - Frank A Ferrone
- Department of Physics, Drexel University, Philadelphia, Pennsylvania.
| |
Collapse
|
14
|
Screen "play" for drug discovery. Proc Natl Acad Sci U S A 2022; 119:e2215625119. [PMID: 36264843 PMCID: PMC9659400 DOI: 10.1073/pnas.2215625119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
15
|
Eaton WA. Drug discovery by a basic research scientist. Front Mol Biosci 2022; 9:1062346. [DOI: 10.3389/fmolb.2022.1062346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
I was fortunate to do my military service during the Vietnam era as a medical officer at the National Institutes of Health (NIH) in Bethesda, Maryland. My first research at NIH was concerned with making a variety of optical measurements on nucleic acid bases and proteins, including single crystal spectra in linearly polarized light and near infrared circular dichroism, interpreting the spectra using molecular orbital and crystal field theories. What I do now is drug discovery, a field at the opposite end of the scientific spectrum. This article gives a brief account of my transition from spectroscopy to sickle cell hemoglobin polymerization to protein folding to drug discovery for treating sickle cell disease. My lab recently developed a high throughput assay to screen the 12,657 compounds of the California Institute of Biomedical Research ReFrame drug repurposing library. This is a precious library because the compounds have either been FDA approved or have been tested in clinical trials. Since the 1970s numerous agents have been reported in the literature to inhibit HbS polymerization and/or sickling with only one successful drug, hydroxyurea, and another of dubious value, voxelotor, even though it has been approved by the FDA. Our screen has discovered 106 anti-sickling agents in the ReFrame compound library. We estimate that as many as 21 of these compounds could become oral drugs for treating sickle cell disease because they inhibit at concentrations typical of the free concentrations of oral drugs in human serum.
Collapse
|