1
|
Hassler JF, Lawson M, Arroyo EC, Bates FS, Hackel BJ, Lodge TP. Discovery of Kinetic Trapping of Poloxamers inside Liposomes via Thermal Treatment. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:14263-14274. [PMID: 37755825 PMCID: PMC10853007 DOI: 10.1021/acs.langmuir.3c01499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Poloxamers, a class of biocompatible, commercially available amphiphilic block polymers (ABPs) comprising poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO) blocks, interact with phospholipid bilayers, resulting in altered mechanical and surface properties. These block copolymers are useful in a variety of applications including therapeutics for Duchenne muscular dystrophy, as cell membrane stabilizers, and for drug delivery, as liposome surface modifying agents. Hydrogen bonding between water and oxygen atoms in PEO and PPO units results in thermoresponsive behavior because the bound water shell around both blocks dehydrates as the temperature increases. This motivated an investigation of poloxamer-lipid bilayer interactions as a function of temperature and thermal history. In this study, we applied pulsed-field-gradient NMR spectroscopy to measure the fraction of chains bound to 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) liposomes between 10 and 50 °C. We measured an (11 ± 3)-fold increase in binding affinity at 37 °C relative to 27 °C. Moreover, following incubation at 37 °C, it takes weeks for the system to re-equilibrate at 25 °C. Such slow desorption kinetics suggests that at elevated temperatures polymer chains can pass through the bilayer and access the interior of the liposomes, a mechanism that is inaccessible at lower temperatures. We propose a molecular mechanism to explain this effect, which could have important ramifications on the cellular distribution of ABPs and could be exploited to modulate the mechanical and surface properties of liposomes and cell membranes.
Collapse
|
2
|
Bez Batti Angulski A, Hosny N, Cohen H, Martin AA, Hahn D, Bauer J, Metzger JM. Duchenne muscular dystrophy: disease mechanism and therapeutic strategies. Front Physiol 2023; 14:1183101. [PMID: 37435300 PMCID: PMC10330733 DOI: 10.3389/fphys.2023.1183101] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and ultimately fatal disease of skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. The identification of the dystrophin gene as central to DMD pathogenesis has led to the understanding of the muscle membrane and the proteins involved in membrane stability as the focal point of the disease. The lessons learned from decades of research in human genetics, biochemistry, and physiology have culminated in establishing the myriad functionalities of dystrophin in striated muscle biology. Here, we review the pathophysiological basis of DMD and discuss recent progress toward the development of therapeutic strategies for DMD that are currently close to or are in human clinical trials. The first section of the review focuses on DMD and the mechanisms contributing to membrane instability, inflammation, and fibrosis. The second section discusses therapeutic strategies currently used to treat DMD. This includes a focus on outlining the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, repair, and/or a range of dystrophin-independent approaches. The final section highlights the different therapeutic strategies for DMD currently in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
3
|
Hahn D, Quick JD, Thompson BR, Crabtree A, Hackel BJ, Bates FS, Metzger JM. Rapid restitution of contractile dysfunction by synthetic copolymers in dystrophin-deficient single live skeletal muscle fibers. Skelet Muscle 2023; 13:9. [PMID: 37208786 PMCID: PMC10197332 DOI: 10.1186/s13395-023-00318-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by the lack of dystrophin, a cytoskeletal protein essential for the preservation of the structural integrity of the muscle cell membrane. DMD patients develop severe skeletal muscle weakness, degeneration, and early death. We tested here amphiphilic synthetic membrane stabilizers in mdx skeletal muscle fibers (flexor digitorum brevis; FDB) to determine their effectiveness in restoring contractile function in dystrophin-deficient live skeletal muscle fibers. After isolating FDB fibers via enzymatic digestion and trituration from thirty-three adult male mice (9 C57BL10, 24 mdx), these were plated on a laminin-coated coverslip and treated with poloxamer 188 (P188; PEO75-PPO30-PEO75; 8400 g/mol), architecturally inverted triblock (PPO15-PEO200-PPO15, 10,700 g/mol), and diblock (PEO75-PPO16-C4, 4200 g/mol) copolymers. We assessed the twitch kinetics of sarcomere length (SL) and intracellular Ca2+ transient by Fura-2AM by field stimulation (25 V, 0.2 Hz, 25 °C). Twitch contraction peak SL shortening of mdx FDB fibers was markedly depressed to 30% of the dystrophin-replete control FDB fibers from C57BL10 (P < 0.001). Compared to vehicle-treated mdx FDB fibers, copolymer treatment robustly and rapidly restored the twitch peak SL shortening (all P < 0.05) by P188 (15 μM = + 110%, 150 μM = + 220%), diblock (15 μM = + 50%, 150 μM = + 50%), and inverted triblock copolymer (15 μM = + 180%, 150 μM = + 90%). Twitch peak Ca2+ transient from mdx FDB fibers was also depressed compared to C57BL10 FDB fibers (P < 0.001). P188 and inverted triblock copolymer treatment of mdx FDB fibers increased the twitch peak Ca2+ transient (P < 0.001). This study shows synthetic block copolymers with varied architectures can rapidly and highly effectively enhance contractile function in live dystrophin-deficient skeletal muscle fibers.
Collapse
Affiliation(s)
- Dongwoo Hahn
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Joseph D Quick
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Brian R Thompson
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Adelyn Crabtree
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Benjamin J Hackel
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Frank S Bates
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
4
|
Lowe M, Bambhroliya Z, Patel H, Patel VJ, Vudugula SA, Cheruvu NP, Raza S, Okunlola OI. Emerging Therapies for the Management of Pain and Vaso-Occlusive Crises in Patients With Sickle Cell Disease: A Systematic Review of Randomized Controlled Trials. Cureus 2023; 15:e38014. [PMID: 37223201 PMCID: PMC10204617 DOI: 10.7759/cureus.38014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 04/23/2023] [Indexed: 05/25/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited disorder that impairs red blood cells (RBCs) and disrupts the delivery of oxygen to tissues. There is currently no cure. Symptoms can appear as early as six months of age and include anemia, acute episodes of pain, swelling, infections, delayed growth, and vision problems. A growing number of therapies are being investigated for reducing these episodes of pain, also known as vaso-occlusive crises (VOCs). The research literature evidence, however, currently includes far more approaches that have not shown superiority versus placebo than ones that have been proven effective. The purpose of this systematic review is to evaluate the body of randomized controlled trials (RCTs) to determine the quality of support for and against the use of a variety of current and emerging therapies for treading SCD VOCs. Several important new papers have emerged since previous systematic reviews with similar objectives were published. This review was conducted according to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines and focused on PubMed exclusively. Only RCTs were sought, and no other filters, except for a five-year historical timeline cut-off, were used. Of the 46 publications that were returned in response to the query, 18 were ultimately accepted as meeting the pre-established inclusion criteria. The Cochrane risk-of-bias tool was utilized as a quality assessment measure, and the GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) framework was used to assess the certainty of the evidence. Among the included publications, five out of 18 featured positive results with superiority and statistical significance versus placebo for either reduction in pain score or number/duration of VOCs. The approaches featured therapies ranging from de novo molecules to currently available drugs approved for other indications to naturally occurring metabolites such as amino acids and vitamins. A single therapy, arginine, was supported for both clinical endpoints: pain score reduction and shortened VOC duration. Currently, two therapies are approved by the United States Food and Drug Administration (FDA) and are commercially available (crizanlizumab, ADAKVEO and L-glutamine, Endari). All other therapies are investigational only in nature. Several studies included measurement of biomarker endpoints as well as clinical outcomes. Generally, beneficial outcomes related to improving biomarker levels did not also translate into statistically significant reduction of pain scores or number/duration of VOCs. While measuring biomarkers may contribute to the understanding of pathophysiology, it does not appear to directly offer predictive value toward treatment success clinically. It can be concluded that there exists a specific opportunity to design, fund, and execute investigations that both compare emerging and existing therapies versus one another and compare combinational therapies versus placebo.
Collapse
Affiliation(s)
- Michael Lowe
- Seeking Anesthesiology, Ross University School of Medicine, Fort Lauderdale, USA
| | - Zarna Bambhroliya
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Hesha Patel
- Internal Medicine, Capital Health Medical Center, New Jersey, USA
| | - Vishva J Patel
- Medicine, Gujarat Medical Education and Research Society (GMERS) Medical College, Vadodara, IND
| | | | | | - Shafaat Raza
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | |
Collapse
|
5
|
Poloxamer 188 as surfactant in biological formulations - An alternative for polysorbate 20/80? Int J Pharm 2022; 620:121706. [PMID: 35367584 DOI: 10.1016/j.ijpharm.2022.121706] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/05/2022] [Accepted: 03/26/2022] [Indexed: 01/25/2023]
Abstract
Surfactants are used to stabilize biologics. Particularly, polysorbates (Tween® 20 and Tween® 80) dominate the group of surfactants in protein and especially antibody drug products. Since decades drug developers rely on the ethoxylated sorbitan fatty acid ester mixtures to stabilize sensitive molecules such as proteins. Reasons are (i) excellent stabilizing properties, and (ii) well recognized safety and tolerability profile of these polysorbates in humans, especially for parenteral applications. However, over the past decade concerns regarding the stability of these two polysorbates were raised. The search of alternatives with preferably less reservations concerning degradation and product quality reducing issues leads, among others, to poloxamer 188 (e.g. Kolliphor® P188), a nonionic triblock-copolymer surfactant. This review sums up our current knowledge related to the characterization and physico-chemical properties of poloxamer 188, its analytics and stability properties for biological formulations. Furthermore, the advantages and disadvantages as a suitable polysorbate-alternative for the stabilization of biologics are discussed.
Collapse
|
6
|
Jayaraman MS, Graham K, Unger EC. Injectable oxygenation therapeutics: evaluating the oxygen delivery efficacy of artificial oxygen carriers and kosmotropes in vitro. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:317-324. [PMID: 33739901 DOI: 10.1080/21691401.2021.1879103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/17/2021] [Indexed: 10/21/2022]
Abstract
The aim of this paper was to utilise an existing in vitro setup to quantify the oxygen offloading capabilities of two different subsets of injectable oxygenation therapeutics: (1) artificial oxygen carriers (AOCs), which bind or dissolve oxygen and act as transport vectors, and (2) kosmotropes, which increase water hydrogen bonding and thereby decrease the resistance to oxygen movement caused by the blood plasma. Dodecafluoropentane emulsion (DDFPe) was chosen to represent the AOC subset while trans sodium crocetinate (TSC) was selected to represent the kosmotrope subset. PEG-Telomer-B (PTB), the surfactant utilised to encapsulate DDFP in emulsion form, was also tested to determine whether it affected the oxygen transport ability of DDFPe. The in vitro set-up was used to simulate a semi closed-loop circulatory system, in which oxygen could be delivered from the lungs to hypoxic tissues. Results of this study showed that (1) 0.5 ml of a PFC outperformed 6.25 ml of a kosmotrope in a controlled, in vitro setting and (2) that PTB and sucrose do not contribute to the overall oxygen transportation efficacy of DDFPe. These results could be therapeutically beneficial to ongoing and future pre-clinical and clinical studies involving various oxygenation agents.
Collapse
|
7
|
Casella JF, Kronsberg SS, Gorney RT. Poloxamer 188 vs Placebo for Painful Vaso-occlusive Episodes in Children and Adults With Sickle Cell Disease-Reply. JAMA 2021; 326:975-976. [PMID: 34519804 DOI: 10.1001/jama.2021.11103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- James F Casella
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rebecca T Gorney
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Lu M, Kanne CK, Reddington RC, Lezzar DL, Sheehan VA, Shevkoplyas SS. Concurrent Assessment of Deformability and Adhesiveness of Sickle Red Blood Cells by Measuring Perfusion of an Adhesive Artificial Microvascular Network. Front Physiol 2021; 12:633080. [PMID: 33995119 PMCID: PMC8113687 DOI: 10.3389/fphys.2021.633080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Biomarker development is a key clinical research need in sickle cell disease (SCD). Hemorheological parameters are excellent candidates as abnormal red blood cell (RBC) rheology plays a critical role in SCD pathophysiology. Here we describe a microfluidic device capable of evaluating RBC deformability and adhesiveness concurrently, by measuring their effect on perfusion of an artificial microvascular network (AMVN) that combines microchannels small enough to require RBC deformation, and laminin (LN) coating on channel walls to model intravascular adhesion. Each AMVN device consists of three identical capillary networks, which can be coated with LN (adhesive) or left uncoated (non-adhesive) independently. The perfusion rate for sickle RBCs in the LN-coated networks (0.18 ± 0.02 nL/s) was significantly slower than in non-adhesive networks (0.20 ± 0.02 nL/s), and both were significantly slower than the perfusion rate for normal RBCs in the LN-coated networks (0.22 ± 0.01 nL/s). Importantly, there was no overlap between the ranges of perfusion rates obtained for sickle and normal RBC samples in the LN-coated networks. Interestingly, treatment with poloxamer 188 decreased the perfusion rate for sickle RBCs in LN-coated networks in a dose-dependent manner, contrary to previous studies with conventional assays, but in agreement with the latest clinical trial which showed no clinical benefit. Overall, these findings suggest the potential utility of the adhesive AMVN device for evaluating the effect of novel curative and palliative therapies on the hemorheological status of SCD patients during clinical trials and in post-market clinical practice.
Collapse
Affiliation(s)
- Madeleine Lu
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Celeste K Kanne
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Riley C Reddington
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Dalia L Lezzar
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Vivien A Sheehan
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Sergey S Shevkoplyas
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
9
|
Casella JF, Barton BA, Kanter J, Black LV, Majumdar S, Inati A, Wali Y, Drachtman RA, Abboud MR, Kilinc Y, Fuh BR, Al-Khabori MK, Takemoto CM, Salman E, Sarnaik SA, Shah N, Morris CR, Keates-Baleeiro J, Raj A, Alvarez OA, Hsu LL, Thompson AA, Sisler IY, Pace BS, Noronha SA, Lasky JL, de Julian EC, Godder K, Thornburg CD, Kamberos NL, Nuss R, Marsh AM, Owen WC, Schaefer A, Tebbi CK, Chantrain CF, Cohen DE, Karakas Z, Piccone CM, George A, Fixler JM, Singleton TC, Moulton T, Quinn CT, de Castro Lobo CL, Almomen AM, Goyal-Khemka M, Maes P, Emanuele M, Gorney RT, Padgett CS, Parsley E, Kronsberg SS, Kato GJ, Gladwin MT. Effect of Poloxamer 188 vs Placebo on Painful Vaso-Occlusive Episodes in Children and Adults With Sickle Cell Disease: A Randomized Clinical Trial. JAMA 2021; 325:1513-1523. [PMID: 33877274 PMCID: PMC8058640 DOI: 10.1001/jama.2021.3414] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPORTANCE Although effective agents are available to prevent painful vaso-occlusive episodes of sickle cell disease (SCD), there are no disease-modifying therapies for ongoing painful vaso-occlusive episodes; treatment remains supportive. A previous phase 3 trial of poloxamer 188 reported shortened duration of painful vaso-occlusive episodes in SCD, particularly in children and participants treated with hydroxyurea. OBJECTIVE To reassess the efficacy of poloxamer 188 for vaso-occlusive episodes. DESIGN, SETTING, AND PARTICIPANTS Phase 3, randomized, double-blind, placebo-controlled, multicenter, international trial conducted from May 2013 to February 2016 that included 66 hospitals in 12 countries and 60 cities; 388 individuals with SCD (hemoglobin SS, SC, S-β0 thalassemia, or S-β+ thalassemia disease) aged 4 to 65 years with acute moderate to severe pain typical of painful vaso-occlusive episodes requiring hospitalization were included. INTERVENTIONS A 1-hour 100-mg/kg loading dose of poloxamer 188 intravenously followed by a 12-hour to 48-hour 30-mg/kg/h continuous infusion (n = 194) or placebo (n = 194). MAIN OUTCOMES AND MEASURES Time in hours from randomization to the last dose of parenteral opioids among all participants and among those younger than 16 years as a separate subgroup. RESULTS Of 437 participants assessed for eligibility, 388 were randomized (mean age, 15.2 years; 176 [45.4%] female), the primary outcome was available for 384 (99.0%), 15-day follow-up contacts were available for 357 (92.0%), and 30-day follow-up contacts were available for 368 (94.8%). There was no significant difference between the groups for the mean time to last dose of parenteral opioids (81.8 h for the poloxamer 188 group vs 77.8 h for the placebo group; difference, 4.0 h [95% CI, -7.8 to 15.7]; geometric mean ratio, 1.2 [95% CI, 1.0-1.5]; P = .09). Based on a significant interaction of age and treatment (P = .01), there was a treatment difference in time from randomization to last administration of parenteral opioids for participants younger than 16 years (88.7 h in the poloxamer 188 group vs 71.9 h in the placebo group; difference, 16.8 h [95% CI, 1.7-32.0]; geometric mean ratio, 1.4 [95% CI, 1.1-1.8]; P = .008). Adverse events that were more common in the poloxamer 188 group than the placebo group included hyperbilirubinemia (12.7% vs 5.2%); those more common in the placebo group included hypoxia (12.0% vs 5.3%). CONCLUSIONS AND RELEVANCE Among children and adults with SCD, poloxamer 188 did not significantly shorten time to last dose of parenteral opioids during vaso-occlusive episodes. These findings do not support the use of poloxamer 188 for vaso-occlusive episodes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01737814.
Collapse
Affiliation(s)
- James F. Casella
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Julie Kanter
- Medical University of South Carolina, Charleston
- University of Alabama at Birmingham, Birmingham
| | - L. Vandy Black
- Our Lady of the Lake Regional Medical Center, Baton Rouge, Louisiana
- University of Florida College of Medicine, Gainesville
| | - Suvankar Majumdar
- University of Mississippi Medical Center, Jackson
- Children’s National Hospital, Washington, DC
| | - Adlette Inati
- Lebanese American University, Byblos and Beirut, Lebanon
- Nini Hospital, Tripoli, Lebanon
| | | | | | | | - Yurdanur Kilinc
- Çukurova University Medical Faculty Balcali Hospital, University of Çukurova, Adana, Turkey
| | - Beng R. Fuh
- East Carolina University, Greenville, North Carolina
| | | | - Clifford M. Takemoto
- Johns Hopkins University School of Medicine, Baltimore, Maryland
- St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Emad Salman
- Golisano Children’s Hospital of Southwest Florida, Ft Myers
| | - Sharada A. Sarnaik
- Wayne State University School of Medicine, Detroit, Michigan
- Children’s Hospital of Michigan, Detroit
| | - Nirmish Shah
- Duke University School of Medicine, Durham, North Carolina
| | - Claudia R. Morris
- Emory University School of Medicine, Atlanta, Georgia
- Children’s Healthcare of Atlanta, Atlanta, Georgia
| | | | - Ashok Raj
- University of Louisville/Norton Children’s Hospital, Louisville, Kentucky
| | | | | | - Alexis A. Thompson
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Evanston, Illinois
| | - India Y. Sisler
- Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond
| | | | - Suzie A. Noronha
- University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital at University of Rochester Medical Center, Rochester, New York
| | - Joseph L. Lasky
- Harbor-UCLA Medical Center, Torrance, California
- Cure 4 The Kids Foundation, Las Vegas, Nevada
| | - Elena Cela de Julian
- Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Courtney Dawn Thornburg
- Rady Children’s Hospital - San Diego, San Diego, California
- UC San Diego School of Medicine, La Jolla, California
| | - Natalie L. Kamberos
- University of Iowa Children’s Hospital, Iowa City
- Loyola University Medical Center, Maywood, Illinois
| | - Rachelle Nuss
- Children’s Hospital Colorado, University of Colorado, Aurora
| | - Anne M. Marsh
- UCSF Benioff Children’s Hospital Oakland (UBCHO), Oakland, California
- University of Wisconsin–Madison, Madison
| | - William C. Owen
- Children’s Hospital of the King’s Daughters, Norfolk, Virginia
| | - Anne Schaefer
- Joe DiMaggio Children’s Hospital, Hollywood, Florida
| | | | | | - Debra E. Cohen
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Studer Family Children’s Hospital Ascension Sacred Heart, University of Florida, Pensacola
| | - Zeynep Karakas
- Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Connie M. Piccone
- Rainbow Babies and Children’s Hospital, Cleveland, Ohio
- Carle Foundation Hospital, Urbana, Illinois
| | - Alex George
- Baylor College of Medicine, Houston, Texas
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jason M. Fixler
- The Herman and Walter Samuelson Children’s Hospital at Sinai, Baltimore, Maryland
| | - Tammuella C. Singleton
- Tulane University, New Orleans, Louisiana
- Mississippi Center for Advanced Medicine, Slidell, Louisiana
| | - Thomas Moulton
- Bronx-Lebanon Hospital, Bronx, New York City, New York
- Bayer Pharmaceuticals, Whippany, New Jersey
| | | | | | - Abdulkareem M. Almomen
- Blood and Cancer Center, King Khalid University Hospital (KKUH), King Saud University Medical City, Riyadh, Saudi Arabia
| | - Meenakshi Goyal-Khemka
- Phoenix Children’s Hospital, Phoenix, Arizona
- Rutgers Cancer Institute of New Jersey, New Brunswick
| | - Philip Maes
- University Hospital of Antwerp (UZA), Edegem, Belgium
| | - Marty Emanuele
- Visgenx, San Diego, California
- Mast Therapeutics Inc, San Diego, California
| | | | - Claire S. Padgett
- Mast Therapeutics Inc, San Diego, California
- Sanifit Therapeutics, San Diego, California
| | - Ed Parsley
- Mast Therapeutics Inc, San Diego, California
- Biotechnology, San Diego, California
| | | | - Gregory J. Kato
- CSL Behring, King of Prussia, Pennsylvania
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mark T. Gladwin
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Hsieh PC, Kuo CY, Wu CP, Yue CT, Peng CK, Huang KL, Lan CC. Nonionic surfactant attenuates acute lung injury by restoring epithelial integrity and alveolar fluid clearance. Int J Med Sci 2021; 18:1363-1374. [PMID: 33628092 PMCID: PMC7893557 DOI: 10.7150/ijms.51905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/06/2021] [Indexed: 01/09/2023] Open
Abstract
Introduction: Acute lung injury (ALI) has a great impact and a high mortality rate in intensive care units (ICUs). Excessive air may enter the lungs, causing pulmonary air embolism (AE)-induced ALI. Some invasive iatrogenic procedures cause pulmonary AE-induced ALI, with the presentation of severe inflammatory reactions, hypoxia, and pulmonary hypertension. Pulmonary surfactants are vital in the lungs to reduce the surface tension and inflammation. Nonionic surfactants (NIS) are a kind of surfactants without electric charge on their hydrophilic parts. Studies on NIS in AE-induced ALI are limited. We aimed to study the protective effects and mechanisms of NIS in AE-induced ALI. Materials and methods: Five different groups (n = 6 in each group) were created: sham, AE, AE + NIS pretreatment (0.5 mg/kg), AE + NIS pretreatment (1 mg/kg), and AE + post-AE NIS (1 mg/kg). AE-induced ALI was introduced by the infusion of air via the pulmonary artery. Aerosolized NIS were administered via tracheostomy. Results: Pulmonary AE-induced ALI showed destruction of the alveolar cell integrity with increased pulmonary microvascular permeability, pulmonary vascular resistance, pulmonary edema, and lung inflammation. The activation of nuclear factor-κB (NF-κB) increased the expression of pro-inflammatory cytokines, and sodium-potassium-chloride co-transporter isoform 1 (NKCC1). The pretreatment with NIS (1 mg/kg) prominently maintained the integrity of the epithelial lining and suppressed the expression of NF-κB, pro-inflammatory cytokines, and NKCC1, subsequently reducing AE-induced ALI. Conclusions: NIS maintained the integrity of the epithelial lining and suppressed the expression of NF-κB, pro-inflammatory cytokines, and NKCC1, thereby reducing hyperpermeability, pulmonary edema, and inflammation in ALI.
Collapse
Affiliation(s)
- Po-Chun Hsieh
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed International Hospital, Tao-Yuan City, Taiwan
| | - Chung-Tai Yue
- Department of Anatomic Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Department of Pathology, Buddhist Tzu Chi University, Hualien, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary Medicine, Tri-Service General Hospital, Taipei, Taiwan; Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Pulmonary Medicine, Tri-Service General Hospital, Taipei, Taiwan; Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chou-Chin Lan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation; School of Medicine, Tzu-Chi University, Hualien, Taiwan
| |
Collapse
|
11
|
Ballas SK. The Evolving Pharmacotherapeutic Landscape for the Treatment of Sickle Cell Disease. Mediterr J Hematol Infect Dis 2020; 12:e2020010. [PMID: 31934320 PMCID: PMC6951351 DOI: 10.4084/mjhid.2020.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Sickle cell disease (SCD) is an extremely heterogeneous disease that has been associated with global morbidity and early mortality. More effective and inexpensive therapies are needed. During the last five years, the landscape of the pharmacotherapy of SCD has changed dramatically. Currently, 54 drugs have been used or under consideration to use for the treatment of SCD. These fall into 3 categories: the first category includes the four drugs (Hydroxyurea, L-Glutamine, Crizanlizumab tmca and Voxelotor) that have been approved by the United States Food and Drug Administration (FDA) based on successful clinical trials. The second category includes 22 drugs that failed, discontinued or terminated for now and the third category includes 28 drugs that are actively being considered for the treatment of SCD. Crizanlizumab and Voxelotor are included in the first and third categories because they have been used in more than one trial. New therapies targeting multiple pathways in the complex pathophysiology of SCD have been achieved or are under continued investigation. The emerging trend seems to be the use of multimodal drugs (i.e. drugs that have different mechanisms of action) to treat SCD similar to the use of multiple chemotherapeutic agents to treat cancer.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
12
|
Ansari J, Gavins FNE. Ischemia-Reperfusion Injury in Sickle Cell Disease: From Basics to Therapeutics. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:706-718. [PMID: 30904156 DOI: 10.1016/j.ajpath.2018.12.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/17/2018] [Accepted: 12/07/2018] [Indexed: 12/17/2022]
Abstract
Sickle cell disease (SCD) is one of the most common hereditary hemoglobinopathies worldwide, affecting almost 400,000 newborns globally each year. It is characterized by chronic hemolytic anemia and endothelial dysfunction, resulting in a constant state of disruption of the vascular system and leading to recurrent episodes of ischemia-reperfusion injury (I/RI) to multiple organ systems. I/RI is a fundamental vascular pathobiological paradigm and contributes to morbidity and mortality in a wide range of conditions, including myocardial infarction, stroke, acute kidney injury, and transplantation. I/RI is characterized by an initial restriction of blood supply to an organ, which can lead to ischemia, followed by the subsequent restoration of perfusion and concomitant reoxygenation. Recent advances in the pathophysiology of SCD have led to an understanding that many of the consequences of this disease can be explained by mechanisms associated with I/RI. The following review focuses on the evolving pathobiology of SCD, how various complications of SCD can be attributed to I/RI, and the role of timely therapeutic intervention(s) based on targeting mediators or pathways that influence I/R insult.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana
| | - Felicity N E Gavins
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana.
| |
Collapse
|
13
|
Houang EM, Bartos J, Hackel BJ, Lodge TP, Yannopoulos D, Bates FS, Metzger JM. Cardiac Muscle Membrane Stabilization in Myocardial Reperfusion Injury. JACC Basic Transl Sci 2019; 4:275-287. [PMID: 31061929 PMCID: PMC6488758 DOI: 10.1016/j.jacbts.2019.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/11/2019] [Accepted: 01/26/2019] [Indexed: 12/11/2022]
Abstract
The phospholipid bilayer membrane that surrounds each cell in the body represents the first and last line of defense for preserving overall cell viability. In several forms of cardiac and skeletal muscle disease, deficits in the integrity of the muscle membrane play a central role in disease pathogenesis. In Duchenne muscular dystrophy, an inherited and uniformly fatal disease of progressive muscle deterioration, muscle membrane instability is the primary cause of disease, including significant heart disease, for which there is no cure or highly effective treatment. Further, in multiple clinical forms of myocardial ischemia-reperfusion injury, the cardiac sarcolemma is damaged and this plays a key role in disease etiology. In this review, cardiac muscle membrane stability is addressed, with a focus on synthetic block copolymers as a unique chemical-based approach to stabilize damaged muscle membranes. Recent advances using clinically relevant small and large animal models of heart disease are discussed. In addition, mechanistic insights into the copolymer-muscle membrane interface, featuring atomistic, molecular, and physiological structure-function approaches are highlighted. Collectively, muscle membrane instability contributes significantly to morbidity and mortality in prominent acquired and inherited heart diseases. In this context, chemical-based muscle membrane stabilizers provide a novel therapeutic approach for a myriad of heart diseases wherein the integrity of the cardiac muscle membrane is at risk.
Collapse
Affiliation(s)
- Evelyne M. Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jason Bartos
- Department of Medicine-Cardiovascular Division, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Timothy P. Lodge
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Demetris Yannopoulos
- Department of Medicine-Cardiovascular Division, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
14
|
Houang EM, Sham YY, Bates FS, Metzger JM. Muscle membrane integrity in Duchenne muscular dystrophy: recent advances in copolymer-based muscle membrane stabilizers. Skelet Muscle 2018; 8:31. [PMID: 30305165 PMCID: PMC6180502 DOI: 10.1186/s13395-018-0177-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023] Open
Abstract
The scientific premise, design, and structure-function analysis of chemical-based muscle membrane stabilizing block copolymers are reviewed here for applications in striated muscle membrane injury. Synthetic block copolymers have a rich history and wide array of applications from industry to biology. Potential for discovery is enabled by a large chemical space for block copolymers, including modifications in block copolymer mass, composition, and molecular architecture. Collectively, this presents an impressive chemical landscape to leverage distinct structure-function outcomes. Of particular relevance to biology and medicine, stabilization of damaged phospholipid membranes using amphiphilic block copolymers, classified as poloxamers or pluronics, has been the subject of increasing scientific inquiry. This review focuses on implementing block copolymers to protect fragile muscle membranes against mechanical stress. The review highlights interventions in Duchenne muscular dystrophy, a fatal disease of progressive muscle deterioration owing to marked instability of the striated muscle membrane. Biophysical and chemical engineering advances are presented that delineate and expand upon current understanding of copolymer-lipid membrane interactions and the mechanism of stabilization. The studies presented here serve to underscore the utility of copolymer discovery leading toward the therapeutic application of block copolymers in Duchenne muscular dystrophy and potentially other biomedical applications in which membrane integrity is compromised.
Collapse
Affiliation(s)
- Evelyne M. Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Yuk Y. Sham
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
- University of Minnesota Informatics Institute, MN, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, MN, USA
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, MN, USA
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| |
Collapse
|
15
|
Abstract
BACKGROUND Sickle cell disease causes significant morbidity and mortality and affects the economic and healthcare status of many countries. Yet historically, the disease has not had commensurate outlays of funds that have been aimed at research and development of drugs and treatment procedures for other diseases. METHODS This review examines several treatment modalities and new drugs developed since the late 1990s that have been used to improve outcomes for patients with sickle cell disease. RESULTS Targeted therapies based upon the pathophysiologic mechanisms of sickle cell disease that result in organ dysfunction and painful episodes include hydroxyurea, L-glutamine, crizanlizumab, and other drugs that are currently on the market or are on the verge of becoming available. These agents have the potential to improve survival and quality of life for individuals with sickle cell disease. Also discussed is stem cell transplantation that, to date, is the only curative approach for this disease, as well as the current status of gene therapy. CONCLUSION These examples demonstrate how the current knowledge of sickle cell disease pathophysiology and treatment approaches intersect. Although interest in sickle cell research has blossomed, many more clinical trials need to be initiated and subjected to more strenuous examination and analysis than have been used in the past.
Collapse
Affiliation(s)
- Renée V. Gardner
- Department of Pediatrics, Louisiana State University Health Sciences Center, Children’s Hospital of New Orleans, New Orleans, LA
| |
Collapse
|
16
|
Houang EM, Bates FS, Sham YY, Metzger JM. All-Atom Molecular Dynamics-Based Analysis of Membrane-Stabilizing Copolymer Interactions with Lipid Bilayers Probed under Constant Surface Tensions. J Phys Chem B 2017; 121:10657-10664. [PMID: 29049887 DOI: 10.1021/acs.jpcb.7b08938] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
An all-atom phospholipid bilayer and triblock copolymer model was developed for molecular dynamics (MD) studies. These were performed to investigate the mechanism of interaction between membrane-stabilizing triblock copolymer P188 and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylcholine (POPC) lipid bilayers under applied lateral surface tension (γ) to model membrane mechanical stress. Results showed that P188 insertion is driven by the hydrophobic poly(propylene oxide) (PPO) core and dependent on bilayer area per lipid. Moreover, insertion of P188 increased the bilayer's resistance to mechanical rupture, as observed by a significant increase in the absolute lateral pressure required to disrupt the bilayer. To further investigate the specific chemical features of P188 underlying membrane stabilizer function, a series of MD simulations with triblock copolymers of the same class as P188 but of varying chemical composition and sizes were performed. Results showed that triblock copolymer insertion into the lipid bilayer is dependent on overall copolymer hydrophobicity, with higher copolymer hydrophobicity requiring a reduced bilayer area per lipid ratio for insertion. Further analysis revealed that the effect of copolymer insertion on membrane mechanical integrity was also dependent on hydrophobicity. Here, P188 insertion significantly increased the absolute apparent lateral pressure required to rupture the POPC bilayer, thereby protecting the membrane against mechanical stress. In marked contrast, highly hydrophobic copolymers decreased the lateral pressure necessary for membrane rupture and thus rendering the membrane significantly more susceptible to mechanical stress. These new in silico findings align with recent experimental findings using synthetic lipid bilayers and in muscle cells in vitro and mouse models in vivo. Collectively, these data underscore the importance of PEO-PPO-PEO copolymer chemical composition in copolymer-based muscle membrane stabilization in vitro and in vivo. All-atom modeling with MD simulations holds promise for investigating novel copolymers with enhanced membrane interacting properties.
Collapse
Affiliation(s)
- Evelyne M Houang
- Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Yuk Y Sham
- Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States.,University of Minnesota Informatics Institute , Minneapolis, Minnesota 55455, United States.,Bioinformatics and Computational Biology Program, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Joseph M Metzger
- Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| |
Collapse
|
17
|
A Novel Method of Human Adipose-Derived Stem Cell Isolation with Resultant Increased Cell Yield. Plast Reconstr Surg 2017; 138:983e-996e. [PMID: 27537222 DOI: 10.1097/prs.0000000000002790] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The authors have developed a novel protocol for isolating adipose-derived stem cells from human lipoaspirate. In this study, they compare their new method to a previously published standard protocol. METHODS Human adipose-derived stem cell isolation was performed using two methods to compare cell yield, cell viability, cell proliferation, and regenerative potential. The new and conventional isolation methods differ in two key areas: the collagenase digestion buffer constituents and the use of an orbital shaker. The osteogenic and adipogenic potential of adipose-derived stem cells isolated using both protocols was assessed in vitro, and gene expression analysis was performed. To assess the ability of the isolated cells to generate bone in vivo, the authors created critical-size calvarial defects in mice, which were treated with adipose-derived stem cells loaded onto hydroxyapatite-coated poly(lactic-co-glycolic acid) scaffolds. To test the ability of the isolated cells to enhance adipogenesis, the cells were added to lipoaspirate and placed beneath the scalp of immunocompromised mice. Fat graft volume retention was subsequently assessed by serial computed tomographic volumetric scanning. RESULTS The new method resulted in a 10-fold increased yield of adipose-derived stem cells compared with the conventional method. Cells harvested using the new method demonstrated significantly increased cell viability and proliferation in vitro (p < 0.05). New method cells also demonstrated significantly enhanced osteogenic and adipogenic differentiation capacity in vitro (p < 0.05) in comparison with the conventional method cells. Both cell groups demonstrated equivalent osteogenic and adipogenic regenerative potential in mice. CONCLUSIONS The authors have developed a protocol that maximizes the yield of adipose-derived stem cells derived from lipoaspirate. The new method cells have increased osteogenic and adipogenic potential in vitro and are not inferior to conventional method cells in terms of their ability to generate bone and fat in vivo. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
18
|
Khan SA, Damanhouri G, Ali A, Khan SA, Khan A, Bakillah A, Marouf S, Al Harbi G, Halawani SH, Makki A. Precipitating factors and targeted therapies in combating the perils of sickle cell disease--- A special nutritional consideration. Nutr Metab (Lond) 2016; 13:50. [PMID: 27508000 PMCID: PMC4977632 DOI: 10.1186/s12986-016-0109-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 07/22/2016] [Indexed: 01/19/2023] Open
Abstract
Nutritional research in sickle cell disease has been the focus in recent times owing to not only specific nutritional deficiencies, but also the improvements associated with less painful episodes. Though hydroxyurea remains the drug of choice, certain adverse health effects on long term supplementation makes room for researches of different compounds. Macro and micro nutrient deficiencies, along with vitamins, play an important role in not only meeting the calorific needs, but also reducing clinical complications and growth abnormalities. Symptoms of hyper protein metabolism, increased cell turnover, increased cardiac output, and appetite suppression due to enhanced cytokine production, might give us leads for better understanding of the mechanisms involved. Different nutritional approaches comprising of traditional herbal therapies, antioxidants, flavonoids, vitamins, minerals etc., reducing oxidative stress and blood aggregation, have been tried out to increase the health potential. Nutritional therapies may also serve complementary to the newer therapies using ozone, hematopoietic stem cell transplantation, antifungal medications, erythropoietin etc. Herein we try to present a holistic picture of the different patho-physiological mechanisms, and nutritional strategies adopted.
Collapse
Affiliation(s)
- Shahida A Khan
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ghazi Damanhouri
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ashraf Ali
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Sarah A Khan
- National Brain Research Center, Manesar, Gurgaon, 122051 India
| | - Aziz Khan
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| | - Ahmed Bakillah
- Department of Medicine, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, New York 11203 United State of America (USA)
| | - Samy Marouf
- Department of Hematology, King Fahd Hospital of the Armed forces, Jeddah, Kingdom of Saudi Arabia ; Department of Medical Laboratory, King Fahd Hospital of the Armed forces, Jeddah, Kingdom of Saudi Arabia
| | - Ghazi Al Harbi
- Department of Hematology, Soliman Fakeeh Hospital Jeddah, Jeddah, Kingdom of Saudi Arabia
| | - Saeed H Halawani
- Department of Hematology, Umm Al Qura University, Faculty of Medicine, Makkah, Kingdom of Saudi Arabia
| | - Ahmad Makki
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Kingdom of Saudi Arabia
| |
Collapse
|
19
|
Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood 2016; 127:810-9. [PMID: 26758919 DOI: 10.1182/blood-2015-09-618553] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/21/2015] [Indexed: 01/09/2023] Open
Abstract
Despite Food and Drug Administration (FDA) approval of hydroxyurea to reduce the frequency of vaso-occlusive episodes, sickle cell disease (SCD) has continued to be treated primarily with analgesics for pain relief. However, elucidation of the multiple pathophysiologic mechanisms leading to vaso-occlusion and tissue injury in SCD has now resulted in a burgeoning effort to identify new treatment modalities to prevent or ameliorate the consequences of the disease. Development of new drugs as well as investigation of drugs previously used in other settings have targeted cell adhesion, inflammatory pathways, upregulation of hemoglobin F, hemoglobin polymerization and sickling, coagulation, and platelet activation. Although these efforts have not yet yielded drugs ready for FDA approval, several early studies have been extremely encouraging. Moreover, the marked increase in clinical pharmaceutical research addressing SCD and the new and old drugs in the pipeline make it reasonable to expect that we will soon have new treatments for SCD.
Collapse
|
20
|
Houang EM, Haman KJ, Filareto A, Perlingeiro RC, Bates FS, Lowe DA, Metzger JM. Membrane-stabilizing copolymers confer marked protection to dystrophic skeletal muscle in vivo. Mol Ther Methods Clin Dev 2015; 2:15042. [PMID: 26623440 PMCID: PMC4641511 DOI: 10.1038/mtm.2015.42] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/01/2015] [Accepted: 10/01/2015] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disease of striated muscle deterioration. A unique therapeutic approach for DMD is the use of synthetic membrane stabilizers to protect the fragile dystrophic sarcolemma against contraction-induced mechanical stress. Block copolymer-based membrane stabilizer poloxamer 188 (P188) has been shown to protect the dystrophic myocardium. In comparison, the ability of synthetic membrane stabilizers to protect fragile DMD skeletal muscles has been less clear. Because cardiac and skeletal muscles have distinct structural and functional features, including differences in the mechanism of activation, variance in sarcolemma phospholipid composition, and differences in the magnitude and types of forces generated, we speculated that optimized membrane stabilization could be inherently different. Our objective here is to use principles of pharmacodynamics to evaluate membrane stabilization therapy for DMD skeletal muscles. Results show a dramatic differential effect of membrane stabilization by optimization of pharmacodynamic-guided route of poloxamer delivery. Data show that subcutaneous P188 delivery, but not intravascular or intraperitoneal routes, conferred significant protection to dystrophic limb skeletal muscles undergoing mechanical stress in vivo. In addition, structure-function examination of synthetic membrane stabilizers further underscores the importance of copolymer composition, molecular weight, and dosage in optimization of poloxamer pharmacodynamics in vivo.
Collapse
Affiliation(s)
- Evelyne M Houang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Karen J Haman
- Department of Chemical Engineering and Material Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Antonio Filareto
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rita C Perlingeiro
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Frank S Bates
- Department of Chemical Engineering and Material Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dawn A Lowe
- Rehabilitation Science and Program in Physical Therapy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Conran N. Prospects for early investigational therapies for sickle cell disease. Expert Opin Investig Drugs 2015; 24:595-602. [DOI: 10.1517/13543784.2015.1012292] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
22
|
Muller AJ, Marks JD. Hypoxic ischemic brain injury: Potential therapeutic interventions for the future. Neoreviews 2014; 15:e177-e186. [PMID: 25177211 DOI: 10.1542/neo.15-5-e177] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Perinatal hypoxic-ischemic brain injury is a common problem with potentially devastating impact on neurodevelopmental outcomes. While therapeutic hypothermia, the first available treatment for this disease, reduces the risk of death or major neurodevelopmental disability, the risk of major neurologic morbidity following HI remains significant. Basic research has identified cellular mechanisms that mediate neuronal death. This article reviews the cellular processes induced that lead to brain injury following HI, and identify treatments currently under investigation for potential translation to clinical trials.
Collapse
Affiliation(s)
- Aaron J Muller
- Department of Pediatrics, University of Chicago 900 East 57th Street Chicago IL 60637
| | - Jeremy D Marks
- Department of Neurology, University of Chicago 900 East 57th Street Chicago IL 60637
| |
Collapse
|
23
|
Townsend D, Yasuda S, Metzger J. Cardiomyopathy of Duchenne muscular dystrophy: pathogenesis and prospect of membrane sealants as a new therapeutic approach. Expert Rev Cardiovasc Ther 2014; 5:99-109. [PMID: 17187461 DOI: 10.1586/14779072.5.1.99] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating progressive disease of striated muscle deterioration. This fatal X-linked disorder results from the loss of the protein dystrophin, which in turn causes striated muscle membrane instability. Cardiac dysfunction is a growing problem in patients with DMD, but relatively little is known about the pathophysiology of the dystrophic heart. At present, there is no effective treatment for DMD and the current clinical approaches are primarily supportive in nature. This review will discuss the pathogenesis of DMD in the heart and discuss how these pathogenic processes have led to a new class of agents directed specifically at restoring membrane integrity to dystrophic myocardium. The tri-block poloxamers, specifically poloxamer 188 (P188), are able to stabilize the membranes of dystrophic myocardium in animal models and may offer a new therapeutic approach for cardiac disease in DMD.
Collapse
Affiliation(s)
- DeWayne Townsend
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | |
Collapse
|
24
|
Abstract
Recurrent and unpredictable episodes of vaso-occlusion are the hallmark of sickle cell disease. Symptomatic management and prevention of these events using the fetal hemoglobin-reactivating agent hydroxyurea are currently the mainstay of treatment. Discoveries over the past 2 decades have highlighted the important contributions of various cellular and soluble participants in the vaso-occlusive cascade. The role of these elements and the opportunities for therapeutic intervention are summarized in this review.
Collapse
|
25
|
Shelat PB, Plant LD, Wang JC, Lee E, Marks JD. The membrane-active tri-block copolymer pluronic F-68 profoundly rescues rat hippocampal neurons from oxygen-glucose deprivation-induced death through early inhibition of apoptosis. J Neurosci 2013; 33:12287-99. [PMID: 23884935 PMCID: PMC3721839 DOI: 10.1523/jneurosci.5731-12.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 06/11/2013] [Accepted: 06/14/2013] [Indexed: 01/01/2023] Open
Abstract
Pluronic F-68, an 80% hydrophilic member of the Pluronic family of polyethylene-polypropylene-polyethylene tri-block copolymers, protects non-neuronal cells from traumatic injuries and rescues hippocampal neurons from excitotoxic and oxidative insults. F-68 interacts directly with lipid membranes and restores membrane function after direct membrane damage. Here, we demonstrate the efficacy of Pluronic F-68 in rescuing rat hippocampal neurons from apoptosis after oxygen-glucose deprivation (OGD). OGD progressively decreased neuronal survival over 48 h in a severity-dependent manner, the majority of cell death occurring after 12 h after OGD. Administration of F-68 for 48 h after OGD rescued neurons from death in a dose-dependent manner. At its optimal concentration (30 μm), F-68 rescued all neurons that would have died after the first hour after OGD. This level of rescue persisted when F-68 administration was delayed 12 h after OGD. F-68 did not alter electrophysiological parameters controlling excitability, NMDA receptor-activated currents, or NMDA-induced increases in cytosolic calcium concentrations. However, F-68 treatment prevented phosphatidylserine externalization, caspase activation, loss of mitochondrial membrane potential, and BAX translocation to mitochondria, indicating that F-68 alters apoptotic mechanisms early in the intrinsic pathway of apoptosis. The profound neuronal rescue provided by F-68 after OGD and the high level of efficacy with delayed administration indicate that Pluronic copolymers may provide a novel, membrane-targeted approach to rescuing neurons after brain ischemia. The ability of membrane-active agents to block apoptosis suggests that membranes or their lipid components play prominent roles in injury-induced apoptosis.
Collapse
Affiliation(s)
- Phullara B. Shelat
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Leigh D. Plant
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Janice C. Wang
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Elizabeth Lee
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| | - Jeremy D. Marks
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
26
|
Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2013:362-369. [PMID: 24319205 DOI: 10.1182/asheducation-2013.1.362] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Recurrent and unpredictable episodes of vaso-occlusion are the hallmark of sickle cell disease. Symptomatic management and prevention of these events using the fetal hemoglobin-reactivating agent hydroxyurea are currently the mainstay of treatment. Discoveries over the past 2 decades have highlighted the important contributions of various cellular and soluble participants in the vaso-occlusive cascade. The role of these elements and the opportunities for therapeutic intervention are summarized in this review.
Collapse
|
27
|
Detterich J, Alexy T, Rabai M, Wenby R, Dongelyan A, Coates T, Wood J, Meiselman H. Low-shear red blood cell oxygen transport effectiveness is adversely affected by transfusion and further worsened by deoxygenation in sickle cell disease patients on chronic transfusion therapy. Transfusion 2012; 53:297-305. [PMID: 22882132 DOI: 10.1111/j.1537-2995.2012.03822.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Simple chronic transfusion therapy (CTT) is a mainstay for stroke prophylaxis in sickle cell anemia, but its effects on hemodynamics are poorly characterized. Transfusion improves oxygen-carrying capacity, reducing demands for high cardiac output. While transfusion decreases factors associated with vasoocclusion, including percent hemoglobin (Hb)S, reticulocyte count, and circulating cell-free Hb, it increases blood viscosity, which reduces microvascular flow. The hematocrit-to-viscosity ratio (HVR) is an index of red blood cell oxygen transport effectiveness that varies with shear stress and balances the benefits of improved oxygen capacity to viscosity-mediated impairment of microvascular flow. We hypothesized that transfusion would improve HVR at high shear despite increased blood viscosity, but would decrease HVR at low shear. STUDY DESIGN AND METHODS To test this hypothesis, we examined oxygenated and deoxygenated blood samples from 15 sickle cell patients on CTT immediately before transfusion and again 12 to 120 hours after transfusion. RESULTS Comparable changes in Hb, hematocrit (Hct), reticulocyte count, and HbS with transfusion were observed in all subjects. Viscosity, Hct, and high-shear HVR increased with transfusion while low-shear HVR decreased significantly. CONCLUSION Decreased low-shear HVR suggests impaired oxygen transport to low-flow regions and may explain why some complications of sickle cell anemia are ameliorated by CTT and others may be made worse.
Collapse
Affiliation(s)
- Jon Detterich
- Division of Cardiology, Children's Hospital Los Angeles, California 90027, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ballas SK, Kesen MR, Goldberg MF, Lutty GA, Dampier C, Osunkwo I, Wang WC, Hoppe C, Hagar W, Darbari DS, Malik P. Beyond the definitions of the phenotypic complications of sickle cell disease: an update on management. ScientificWorldJournal 2012; 2012:949535. [PMID: 22924029 PMCID: PMC3415156 DOI: 10.1100/2012/949535] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/01/2012] [Indexed: 12/25/2022] Open
Abstract
The sickle hemoglobin is an abnormal hemoglobin due to point mutation (GAG → GTG) in exon 1 of the β globin gene resulting in the substitution of glutamic acid by valine at position 6 of the β globin polypeptide chain. Although the molecular lesion is a single-point mutation, the sickle gene is pleiotropic in nature causing multiple phenotypic expressions that constitute the various complications of sickle cell disease in general and sickle cell anemia in particular. The disease itself is chronic in nature but many of its complications are acute such as the recurrent acute painful crises (its hallmark), acute chest syndrome, and priapism. These complications vary considerably among patients, in the same patient with time, among countries and with age and sex. To date, there is no well-established consensus among providers on the management of the complications of sickle cell disease due in part to lack of evidence and in part to differences in the experience of providers. It is the aim of this paper to review available current approaches to manage the major complications of sickle cell disease. We hope that this will establish another preliminary forum among providers that may eventually lead the way to better outcomes.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation and Department of Medicine, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Acute chest syndrome (ACS) is a common complication and reason for hospital admission in patients with sickle cell disease (SCD). It is also the most common cause of death in this patient population. Most of the time, the trigger for ACS in an individual patient cannot be identified. However, although infection is the most common identifiable cause for ACS, other important triggers are vaso-occlusive crisis (VOC) and asthma. This comprehensive review will focus on the pathogenesis, clinical characteristics, complications and treatment available to manage ACS. But importantly, this review will highlight new possible etiologies, with the goal of improving oxygenation and, therefore, a reduction in sickling and lung damage in this patient population.
Collapse
Affiliation(s)
- Rabindra N Paul
- Department of Medicine, Division of Hematology/Oncology, Howard University, 2041 Georgia Ave. NW, Washington, DC 20060, USA.
| | | | | | | |
Collapse
|
30
|
Plataki M, Lee YD, Rasmussen DL, Hubmayr RD. Poloxamer 188 facilitates the repair of alveolus resident cells in ventilator-injured lungs. Am J Respir Crit Care Med 2011; 184:939-47. [PMID: 21778295 DOI: 10.1164/rccm.201104-0647oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Wounded alveolus resident cells are identified in human and experimental acute respiratory distress syndrome models. Poloxamer 188 (P188) is an amphiphilic macromolecule shown to have plasma membrane-sealing properties in various cell types. OBJECTIVES To investigate whether P188 (1) protects alveolus resident cells from necrosis and (2) is associated with reduced ventilator-induced lung injury in live rats, isolated perfused rat lungs, and scratch and stretch-wounded alveolar epithelial cells. METHODS Seventy-four live rats and 18 isolated perfused rat lungs were ventilated with injurious or protective strategies while infused with P188 or control solution. Alveolar epithelial cell monolayers were subjected to scratch or stretch wounding in the presence or absence of P188. MEASUREMENTS AND MAIN RESULTS P188 was associated with fewer mortally wounded alveolar cells in live rats and isolated perfused lungs. In vitro, P188 reduced the number of injured and necrotic cells, suggesting that P188 promotes cell repair and renders plasma membranes more resilient to deforming stress. The enhanced cell survival was accompanied by improvement in conventional measures of lung injury (peak airway pressure, wet-to-dry weight ratio) only in the ex vivo-perfused lung preparation and not in the live animal model. CONCLUSIONS P188 facilitates plasma membrane repair in alveolus resident cells, but has no salutary effects on lung mechanics or vascular barrier properties in live animals. This discordance may have pathophysiological significance for the interdependence of different injury mechanisms and therapeutic implications regarding the benefits of prolonging the life of stress-activated cells.
Collapse
Affiliation(s)
- Maria Plataki
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
31
|
Spurney CF. Cardiomyopathy of duchenne muscular dystrophy: Current understanding and future directions. Muscle Nerve 2011; 44:8-19. [DOI: 10.1002/mus.22097] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
32
|
Spurney CF, Guerron AD, Yu Q, Sali A, van der Meulen JH, Hoffman EP, Nagaraju K. Membrane sealant Poloxamer P188 protects against isoproterenol induced cardiomyopathy in dystrophin deficient mice. BMC Cardiovasc Disord 2011; 11:20. [PMID: 21575230 PMCID: PMC3123649 DOI: 10.1186/1471-2261-11-20] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 05/16/2011] [Indexed: 11/15/2022] Open
Abstract
Background Cardiomyopathy in Duchenne muscular dystrophy (DMD) is an increasing cause of death in patients. The absence of dystrophin leads to loss of membrane integrity, cell death and fibrosis in cardiac muscle. Treatment of cardiomyocyte membrane instability could help prevent cardiomyopathy. Methods Three month old female mdx mice were exposed to the β1 receptor agonist isoproterenol subcutaneously and treated with the non-ionic tri-block copolymer Poloxamer P188 (P188) (460 mg/kg/dose i.p. daily). Cardiac function was assessed using high frequency echocardiography. Tissue was evaluated with Evans Blue Dye (EBD) and picrosirius red staining. Results BL10 control mice tolerated 30 mg/kg/day of isoproterenol for 4 weeks while death occurred in mdx mice at 30, 15, 10, 5 and 1 mg/kg/day within 24 hours. Mdx mice tolerated a low dose of 0.5 mg/kg/day. Isoproterenol exposed mdx mice showed significantly increased heart rates (p < 0.02) and cardiac fibrosis (p < 0.01) over 4 weeks compared to unexposed controls. P188 treatment of mdx mice significantly increased heart rate (median 593 vs. 667 bpm; p < 0.001) after 2 weeks and prevented a decrease in cardiac function in isoproterenol exposed mice (Shortening Fraction = 46 ± 6% vs. 35 ± 6%; p = 0.007) after 4 weeks. P188 treated mdx mice did not show significant differences in cardiac fibrosis, but demonstrated significantly increased EBD positive fibers. Conclusions This model suggests that chronic intermittent intraperitoneal P188 treatment can prevent isoproterenol induced cardiomyopathy in dystrophin deficient mdx mice.
Collapse
Affiliation(s)
- Christopher F Spurney
- Children’s National Heart Institute, Division of Cardiology, Children’s National Medical Center, Washington, DC, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Jung J, Choi B, Cho S, Choe S, Ghim J, Lee H, Roh Y, Noh G. Effectiveness, safety, and pharmacokinetic and pharmacodynamic characteristics of microemulsion propofol in patients undergoing elective surgery under total intravenous anaesthesia ‡. Br J Anaesth 2010; 104:563-76. [DOI: 10.1093/bja/aeq040] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
34
|
Abstract
Sickle cell anemia is one of the most common autosomal recessive diseases in the world. Patients with sickle cell anemia have variable penetrance and it is hard to predict the risk and timing of complications. It is characterized by a point mutation in the beta-globin gene (GAG --> GTG) and the production of hemoglobin S. The latter leads to decreased deformability of the red blood cells (RBCs) that adhere to endothelia cells culminating in vascular occlusion and its sequelae of tissue ischemia and organ damage. Moreover, sickled RBCs undergo intravascular hemolysis and accelerated erythropoesis. The hallmarks of this disease are shortened RBC survival and vaso-occlusive crises. For the past ten years, the pathophysiology of this disease has been better elucidated and has led to significant improvements in the standard of care. Vaso-occlusion is now understood to be a complex event that involves abnormal interactions between RBCs, leukocytes, endothelial cells and the coagulation pathways. The field of translational research in sickle cell anemia has expanded greatly and has led to new clinical trials with new therapeutic agents and strategies. In this paper, we review the drugs that are now being investigated in the treatment of sickle cell anemia.
Collapse
|
35
|
Development of a nasal adenovirus-based vaccine: Effect of concentration and formulation on adenovirus stability and infectious titer during actuation from two delivery devices. Vaccine 2010; 28:2137-48. [DOI: 10.1016/j.vaccine.2009.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/10/2009] [Accepted: 12/14/2009] [Indexed: 11/19/2022]
|
36
|
Poloxamer 188 prolongs survival of hypotensive resuscitation and decreases vital tissue injury after full resuscitation. Shock 2010; 32:442-50. [PMID: 19197229 DOI: 10.1097/shk.0b013e31819e13b1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypotensive resuscitation prolongs survival of patients with severe bleeding until they can undergo hemorrhage control. However, its value is limited by continuing ischemic injury. Purified poloxamer 188 (P188), a copolymer with rheological and cytoprotective activities, was known to reduce mortality of hemorrhagic shock when used as an adjunct to full resuscitation with fresh whole blood and crystalloid. Studies were undertaken to determine if it could prolong survival and reduce reperfusion injury during prolonged hypotensive resuscitation when added to the best regimen currently available. Unanesthetized rats were bled to a MAP of 30 mmHg for 30 min under computer control. They then received hypotensive resuscitation with Hextend or Hextend + P188 to maintain a MAP of 60 mmHg until death. Poloxamer 188 improved autoresuscitation, reduced fluid requirements, and increased the survivable duration of hypotensive resuscitation by more than 3 h (P < 0.01). Additional studies assessed tissue damage after shock and hypotensive resuscitation with Hextend followed by full resuscitation with crystalloid. In these studies, P188 blunted the no-reflow phenomenon and largely prevented myocardial injury, pulmonary inflammation, small bowel damage, renal tubular necrosis, hepatic central lobular necrosis, and apoptosis of splenic germinal centers that occurred during full resuscitation. Additional studies demonstrated that P188 increased survival from 0% to 75% in 50% volume-controlled hemorrhage (P < 0.001). Finally, P188 did not increase bleeding in uncontrolled hemorrhage produced by 75% tail amputation. Because P188 prolongs survival, decreases fluid requirements, and reduces tissue damage, it deserves further consideration as an adjunct to hypotensive resuscitation.
Collapse
|
37
|
Ballas SK, Files B, Luchtman-Jones L, Benjamin L, Swerdlow P, Hilliard L, Coates T, Abboud M, Wojtowicz-Praga S, Kuypers FA, Michael Grindel J. Secretory Phospholipase A2Levels in Patients with Sickle Cell Disease and Acute Chest Syndrome. Hemoglobin 2009; 30:165-70. [PMID: 16798640 DOI: 10.1080/03630260600642260] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In a multicenter study (eight centers), we determined secretory phospholipase A(2) (sPLA(2)) levels in patients with sickle cell disease and acute chest syndrome (ACS). The diagnosis of ACS was made according to established criteria. The sPLA2 levels were determined in blood samples collected at baseline (time of diagnosis) and serially thereafter up to day 22-35 follow-up visits. Thirty-four of 43 (80%) patients with ACS had enzyme levels > or =1.00 AU at baseline. The enzyme levels decreased significantly on Days 2 through Days 25-35 after baseline. Nine of 43 (20%) patients had baseline sPLA2 values of <1.00 AU with six of them never exceeding 1.00 AU at any point in time during follow-up. The data indicate that the reliability of sPLA(2( for predicting the development of ACS is not perfect (100%) as was previously reported but occurs in about 80% of the patients.
Collapse
Affiliation(s)
- Samir K Ballas
- Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Fawibe AE. Managing acute chest syndrome of sickle cell disease in an African setting. Trans R Soc Trop Med Hyg 2008; 102:526-31. [PMID: 18455745 DOI: 10.1016/j.trstmh.2008.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Revised: 03/17/2008] [Accepted: 03/19/2008] [Indexed: 11/27/2022] Open
Abstract
Despite the fact that acute chest syndrome contributes immensely to morbidity and mortality in patients with sickle cell anaemia, its exact aetiopathogenesis is very complex and not yet well understood. Therefore, a high index of suspicion is needed in its diagnosis, and appropriate treatment should be commenced as soon as possible to prevent lethal complications of this condition, especially in Nigeria where appropriate diagnostic and therapeutic facilities may not be readily available. This is very important, as it may even develop on hospital admission. There is a need to further investigate preventive measures such as the use of hydroxyurea and the newly introduced Nicosan, especially in those people with recurrent disease, in order to reduce both short- and long-term complications of this syndrome among sickle cell patients in Nigeria.
Collapse
Affiliation(s)
- A E Fawibe
- Department of Internal Medicine, Federal Medical Center, Bida, Niger State, Nigeria.
| |
Collapse
|
40
|
Makis AC, Hatzimichael EC, Stebbing J. The genomics of new drugs in sickle cell disease. Pharmacogenomics 2006; 7:909-17. [PMID: 16981849 DOI: 10.2217/14622416.7.6.909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The quality of life of patients with sickle cell disease in developed countries has improved significantly over the past two decades. Currently available measures to prevent the painful crises and the complications of the disease include the use of penicillin prophylaxis, antipneumonococcal vaccine, folate administration, chronic red cell transfusions in patients with cerebrovascular disease, iron chelating agents, fetal hemoglobin-enhancing agents, such as hydroxyurea, decitabine and butyrate, drugs that augment the endogenous nitric oxide levels and agents that restore red cell dehydration. Sickle cell patients show a broad phenotypic expression and a great variability in treatment response. Genetic association studies, which attempt to link polymorphisms with certain disease phenotypes and drug response, are taking the first steps in aiding individualized therapy in sickle cell patients in order to enhance efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Alexandros C Makis
- Department of Paediatrics, University Hospital of Ioannina, Ioannina, Greece.
| | | | | |
Collapse
|
41
|
Alexy T, Pais E, Armstrong JK, Meiselman HJ, Johnson CS, Fisher TC. Rheologic behavior of sickle and normal red blood cell mixtures in sickle plasma: implications for transfusion therapy. Transfusion 2006; 46:912-8. [PMID: 16734807 DOI: 10.1111/j.1537-2995.2006.00823.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Guidelines for transfusion in sickle cell disease usually define an upper hematocrit (Hct) limit of 0.30 to 0.35 to avoid blood hyperviscosity. In vitro viscosity studies of normal (AA) and sickle (SS) red blood cell (RBC) mixtures in buffer appear to confirm that this Hct limit is optimal for oxygen delivery to vascular beds as judged by the ratio of Hct to viscosity, with this ratio often termed "oxygen or RBC transport effectiveness." In the absence of plasma, however, effects due to RBC-RBC interactions mediated by plasma proteins cannot be assessed. STUDY DESIGNS AND METHODS To investigate the optimal Hct-to-viscosity ratio of RBCs in plasma, the rheologic effects of Hct (0.20-0.40), the proportion of SS RBCs (0-100%), and shear rate (1-1000/sec) for mixtures of oxygenated and deoxygenated SS and AA RBCs were evaluated in sickle plasma at 37 degrees C. RESULTS RBC suspension viscosity was shear-dependent (i.e., viscosity decreased with increasing shear rate) and increased with Hct and proportion of SS RBCs. An "optimal" Hct level (defined as a maximal of the Hct-to-viscosity ratio) was seen only at shear rates above 50/sec. At lower shear rates (e.g., 5/sec), where plasma-mediated RBC-RBC interactions predominate, any increment in Hct was offset by a proportionally greater increase in viscosity, thus leading to a lower Hct-to-viscosity ratio. CONCLUSION These results indicate the importance of plasma-mediated RBC interactions and suggest that the benefits of transfusion may vary depending on local flow rates (i.e., shear rates) and organ-specific hemodynamics.
Collapse
Affiliation(s)
- Tamas Alexy
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|