1
|
Butt H, Sathish S, London E, Lee Johnson T, Essawi K, Leonard A, Tisdale JF, Demirci S. Genome editing strategies for targeted correction of β-globin mutation in sickle cell disease: From bench to bedside. Mol Ther 2025:S1525-0016(25)00221-7. [PMID: 40165374 DOI: 10.1016/j.ymthe.2025.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025] Open
Abstract
Sickle cell disease (SCD) includes a range of genotypes that result in a clinical syndrome, where abnormal red blood cell (RBC) physiology leads to widespread complications affecting nearly every organ system. Treatment strategies for SCD can be broadly categorized into disease-modifying therapies and those aimed toward a cure. Although several disease-modifying drugs have been approved, they do not fully address the complexity and severity of SCD. Recent advances in allogeneic transplantation and autologous gene therapy show promising outcomes in terms of efficacy and safety. While these approaches have improved the lives of many patients, achieving a durable and comprehensive cure for all remains challenging. To address this, gene-editing technologies, including zinc-finger nucleases, TALENs, CRISPR-Cas, base editing, and prime editing, have been explored both ex vivo and in vivo for targeted correction of the β-globin gene (HBB) in SCD. However, direct correction of HBB and its translation from the laboratory to the clinic presents ongoing limitations, with challenges involved in achieving robust mutation-correction efficiency, off-target effects, and high costs of therapies. The optimal strategy for curing SCD remains uncertain, but several promising approaches are emerging. This review touches on past, present, and future developments in HBB correction.
Collapse
Affiliation(s)
- Henna Butt
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Shruti Sathish
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Evan London
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Taylor Lee Johnson
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan 45142, Saudi Arabia
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch (CMTB), National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| |
Collapse
|
2
|
Lewis J, Guilcher GMT, Greenway SC. Reviewing the impact of hydroxyurea on DNA methylation and its potential clinical implications in sickle cell disease. Eur J Haematol 2024; 113:264-272. [PMID: 38831675 DOI: 10.1111/ejh.14247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
Hydroxyurea (HU) is the most common drug therapy for sickle cell disease (SCD). The clinical benefits of HU derive from its upregulation of fetal hemoglobin (HbF), which reduces aggregation of the mutated sickle hemoglobin protein (HbS) and reduces SCD symptoms and complications. However, some individuals do not respond to HU, or stop responding over time. Unfortunately, current understanding of the mechanism of action of HU is limited, hindering the ability of clinicians to identify those patients who will respond to HU and to optimize treatment for those receiving HU. Given that epigenetic modifications are essential to erythropoiesis and HbF expression, we hypothesize that some effects of HU may be mediated by epigenetic modifications, specifically DNA methylation. However, few studies have investigated this possibility and the effects of HU on DNA methylation remain relatively understudied. In this review, we discuss the evidence linking HU treatment to DNA methylation changes and associated gene expression changes, with an emphasis on studies that were performed in individuals with SCD. Overall, although HU can affect DNA methylation, research on these changes and their clinical effects remains limited. Further study is likely to contribute to our understanding of hematopoiesis and benefit patients suffering from SCD.
Collapse
Affiliation(s)
- Jasmine Lewis
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gregory M T Guilcher
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Steven C Greenway
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
3
|
Butt H, Tisdale JF. Gene therapies on the horizon for sickle cell disease: a clinician's perspective. Expert Rev Hematol 2024; 17:555-566. [PMID: 39076056 DOI: 10.1080/17474086.2024.2386366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/20/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Sickle cell disease (SCD) is a monogenic disorder that exerts several detrimental health effects on those affected, ultimately resulting in significant morbidity and early mortality. There are millions of individuals globally impacted by this disease. Research in gene therapy has been growing significantly over the past decade, now with two FDA approved products, aiming to find another cure for this complex disease. AREAS COVERED This perspective article aims to provide a clinician's insight into the current landscape of gene therapies, exploring the novel approaches, clinical advances, and potential impact on the management and prognosis of SCD. A comprehensive literature search encompassing databases such as PubMed, Web of Science and Google Scholar was employed. The search covered literature published from 1980 to 2024, focusing on SCD and curative therapy. EXPERT OPINION After careful evaluation of the risks and benefits associated with the use of gene therapy for affected patients, the need for a cure outweighs the risks associated with treatment in most cases of SCD. With advances in current technologies, gene therapies can increase access to cures for patients with SCD.
Collapse
Affiliation(s)
- Henna Butt
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Condé M, Lespessailles E, Wanneveich M, Allemandou D, Boulain T, Dimitrov G. Effect of nutritional supplementation on bone mineral density in children with sickle cell disease: protocol for an open-label, randomised controlled clinical trial. BMJ Open 2024; 14:e080235. [PMID: 38580373 PMCID: PMC11002356 DOI: 10.1136/bmjopen-2023-080235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/16/2024] [Indexed: 04/07/2024] Open
Abstract
INTRODUCTION Children with sickle cell disease show a significant decrease in bone mineral density, an increase in resting energy expenditure of more than 15%, a decrease in fat and lean mass as well as a significant increase in protein turnover, particularly in bone tissue. This study aims to evaluate the effectiveness of an increase in food intake on bone mineral density and the clinical and biological complications of paediatric sickle cell disease. METHODS AND ANALYSIS The study is designed as an open-label randomised controlled clinical trial conducted in the Paediatrics Unit of the Orléans University Hospital Centre. Participants aged 3-16 years will be randomly divided into two groups: the intervention group will receive oral nutritional supplements (pharmacological nutritional hypercaloric products) while the control group will receive age-appropriate and gender-appropriate nutritional intake during 12 months. Total body less head bone mineral density will be measured at the beginning and the end of the trial. A rigorous nutritional follow-up by weekly 24 hours recall dietary assessment and planned contacts every 6 weeks will be carried out throughout the study. A school absenteeism questionnaire, intended to reflect the patient's school productivity, will be completed by participants and parents every 3 months. Blood samples of each patient of both groups will be stocked at the beginning and at the end of the trial, for future biological trial. Clinical and biological complications will be regularly monitored. ETHICS AND DISSEMINATION The protocol has been approved by the French ethics committee (Comité de Protection des Personnes Sud-Ouest et Outre-Mer 2, Toulouse; approval no: 2-20-092 id9534). Children and their parents will give informed consent to participate in the study before taking part. Results will be disseminated through peer-reviewed journals or international academic conferences. TRIAL REGISTRATION NUMBER NCT04754711.
Collapse
Affiliation(s)
- Mohamed Condé
- Paediatrics, Centre Hospitalier Régional d'Orléans, Orléans, France
| | | | - Mathilde Wanneveich
- Statistics Department, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Delphine Allemandou
- Nutrition Department, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Thierry Boulain
- Medical Intensive Care Unit, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Georges Dimitrov
- Paediatrics, Centre Hospitalier Régional d'Orléans, Orleans, France
- Paediatric Surgery, Centre Hospitalier Régional d'Orléans, Orléans, France
| |
Collapse
|
5
|
Thomson AM, McHugh TA, Oron AP, Teply C, Lonberg N, Vilchis Tella V, Wilner LB, Fuller K, Hagins H, Aboagye RG, Aboye MB, Abu-Gharbieh E, Abu-Zaid A, Addo IY, Ahinkorah BO, Ahmad A, AlRyalat SAS, Amu H, Aravkin AY, Arulappan J, Atout MMW, Badiye AD, Bagherieh S, Banach M, Banakar M, Bardhan M, Barrow A, Bedane DA, Bensenor IM, Bhagavathula AS, Bhardwaj P, Bhardwaj PV, Bhat AN, Bhutta ZA, Bilalaga MM, Bishai JD, Bitaraf S, Boloor A, Butt MH, Chattu VK, Chu DT, Dadras O, Dai X, Danaei B, Dang AK, Demisse FW, Dhimal M, Diaz D, Djalalinia S, Dongarwar D, Elhadi M, Elmonem MA, Esezobor CI, Etaee F, Eyawo O, Fagbamigbe AF, Fatehizadeh A, Force LM, Gardner WM, Ghaffari K, Gill PS, Golechha M, Goleij P, Gupta VK, Hasani H, Hassan TS, Hassen MB, Ibitoye SE, Ikiroma AI, Iwu CCD, James PB, Jayaram S, Jebai R, Jha RP, Joseph N, Kalantar F, Kandel H, Karaye IM, Kassahun WD, Khan IA, Khanmohammadi S, Kisa A, Kompani F, Krishan K, Landires I, Lim SS, Mahajan PB, Mahjoub S, Majeed A, Marasini BP, Meresa HA, Mestrovic T, Minhas S, Misganaw A, Mokdad AH, Monasta L, Mustafa G, Nair TS, Narasimha Swamy S, Nassereldine H, et alThomson AM, McHugh TA, Oron AP, Teply C, Lonberg N, Vilchis Tella V, Wilner LB, Fuller K, Hagins H, Aboagye RG, Aboye MB, Abu-Gharbieh E, Abu-Zaid A, Addo IY, Ahinkorah BO, Ahmad A, AlRyalat SAS, Amu H, Aravkin AY, Arulappan J, Atout MMW, Badiye AD, Bagherieh S, Banach M, Banakar M, Bardhan M, Barrow A, Bedane DA, Bensenor IM, Bhagavathula AS, Bhardwaj P, Bhardwaj PV, Bhat AN, Bhutta ZA, Bilalaga MM, Bishai JD, Bitaraf S, Boloor A, Butt MH, Chattu VK, Chu DT, Dadras O, Dai X, Danaei B, Dang AK, Demisse FW, Dhimal M, Diaz D, Djalalinia S, Dongarwar D, Elhadi M, Elmonem MA, Esezobor CI, Etaee F, Eyawo O, Fagbamigbe AF, Fatehizadeh A, Force LM, Gardner WM, Ghaffari K, Gill PS, Golechha M, Goleij P, Gupta VK, Hasani H, Hassan TS, Hassen MB, Ibitoye SE, Ikiroma AI, Iwu CCD, James PB, Jayaram S, Jebai R, Jha RP, Joseph N, Kalantar F, Kandel H, Karaye IM, Kassahun WD, Khan IA, Khanmohammadi S, Kisa A, Kompani F, Krishan K, Landires I, Lim SS, Mahajan PB, Mahjoub S, Majeed A, Marasini BP, Meresa HA, Mestrovic T, Minhas S, Misganaw A, Mokdad AH, Monasta L, Mustafa G, Nair TS, Narasimha Swamy S, Nassereldine H, Natto ZS, Naveed M, Nayak BP, Noubiap JJ, Noyes T, Nri-ezedi CA, Nwatah VE, Nzoputam CI, Nzoputam OJ, Okonji OC, Onikan AO, Owolabi MO, Patel J, Pati S, Pawar S, Petcu IR, Piel FB, Qattea I, Rahimi M, Rahman M, Rawaf S, Redwan EMM, Rezaei N, Saddik B, Saeed U, Saheb Sharif-Askari F, Samy AM, Schumacher AE, Shaker E, Shetty A, Sibhat MM, Singh JA, Suleman M, Sunuwar DR, Szeto MD, Tamuzi JJLL, Tat NY, Taye BT, Temsah MH, Umair M, Valadan Tahbaz S, Wang C, Wickramasinghe ND, Yigit A, Yiğit V, Yunusa I, Zaman BA, Zangiabadian M, Zheng P, Hay SI, Naghavi M, Murray CJL, Kassebaum NJ. Global, regional, and national prevalence and mortality burden of sickle cell disease, 2000-2021: a systematic analysis from the Global Burden of Disease Study 2021. Lancet Haematol 2023; 10:e585-e599. [PMID: 37331373 PMCID: PMC10390339 DOI: 10.1016/s2352-3026(23)00118-7] [Show More Authors] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Previous global analyses, with known underdiagnosis and single cause per death attribution systems, provide only a small insight into the suspected high population health effect of sickle cell disease. Completed as part of the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2021, this study delivers a comprehensive global assessment of prevalence of sickle cell disease and mortality burden by age and sex for 204 countries and territories from 2000 to 2021. METHODS We estimated cause-specific sickle cell disease mortality using standardised GBD approaches, in which each death is assigned to a single underlying cause, to estimate mortality rates from the International Classification of Diseases (ICD)-coded vital registration, surveillance, and verbal autopsy data. In parallel, our goal was to estimate a more accurate account of sickle cell disease health burden using four types of epidemiological data on sickle cell disease: birth incidence, age-specific prevalence, with-condition mortality (total deaths), and excess mortality (excess deaths). Systematic reviews, supplemented with ICD-coded hospital discharge and insurance claims data, informed this modelling approach. We employed DisMod-MR 2.1 to triangulate between these measures-borrowing strength from predictive covariates and across age, time, and geography-and generated internally consistent estimates of incidence, prevalence, and mortality for three distinct genotypes of sickle cell disease: homozygous sickle cell disease and severe sickle cell β-thalassaemia, sickle-haemoglobin C disease, and mild sickle cell β-thalassaemia. Summing the three models yielded final estimates of incidence at birth, prevalence by age and sex, and total sickle cell disease mortality, the latter of which was compared directly against cause-specific mortality estimates to evaluate differences in mortality burden assessment and implications for the Sustainable Development Goals (SDGs). FINDINGS Between 2000 and 2021, national incidence rates of sickle cell disease were relatively stable, but total births of babies with sickle cell disease increased globally by 13·7% (95% uncertainty interval 11·1-16·5), to 515 000 (425 000-614 000), primarily due to population growth in the Caribbean and western and central sub-Saharan Africa. The number of people living with sickle cell disease globally increased by 41·4% (38·3-44·9), from 5·46 million (4·62-6·45) in 2000 to 7·74 million (6·51-9·2) in 2021. We estimated 34 400 (25 000-45 200) cause-specific all-age deaths globally in 2021, but total sickle cell disease mortality burden was nearly 11-times higher at 376 000 (303 000-467 000). In children younger than 5 years, there were 81 100 (58 800-108 000) deaths, ranking total sickle cell disease mortality as 12th (compared to 40th for cause-specific sickle cell disease mortality) across all causes estimated by the GBD in 2021. INTERPRETATION Our findings show a strikingly high contribution of sickle cell disease to all-cause mortality that is not apparent when each death is assigned to only a single cause. Sickle cell disease mortality burden is highest in children, especially in countries with the greatest under-5 mortality rates. Without comprehensive strategies to address morbidity and mortality associated with sickle cell disease, attainment of SDG 3.1, 3.2, and 3.4 is uncertain. Widespread data gaps and correspondingly high uncertainty in the estimates highlight the urgent need for routine and sustained surveillance efforts, further research to assess the contribution of conditions associated with sickle cell disease, and widespread deployment of evidence-based prevention and treatment for those with sickle cell disease. FUNDING Bill & Melinda Gates Foundation.
Collapse
|
6
|
Inam Z, Tisdale JF, Leonard A. Outcomes and long-term effects of hematopoietic stem cell transplant in sickle cell disease. Expert Rev Hematol 2023; 16:879-903. [PMID: 37800996 DOI: 10.1080/17474086.2023.2268271] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Hematopoietic stem cell transplant (HSCT) is the only readily available curative option for sickle cell disease (SCD). Cure rates following human leukocyte antigen (HLA)-matched related donor HSCT with myeloablative or non-myeloablative conditioning are >90%. Alternative donor sources, including haploidentical donor and autologous with gene therapy, expand donor options but are limited by inferior outcomes, limited data, and/or shorter follow-up and therefore remain experimental. AREAS COVERED Outcomes are improving with time, with donor type and conditioning regimens having the greatest impact on long-term complications. Patients with stable donor engraftment do not experience SCD-related symptoms and have stabilization or improvement of end-organ pathology; however, the long-term effects of curative strategies remain to be fully established and have significant implications in a patient's decision to seek therapy. This review covers currently published literature on HSCT outcomes, including organ-specific outcomes implicated in SCD, as well as long-term effects. EXPERT OPINION HSCT, both allogeneic and autologous gene therapy, in the SCD population reverses the sickle phenotype, prevents further organ damage, can resolve prior organ dysfunction in both pediatric and adult patients. Data support greater success with HSCT at a younger age, thus, curative therapies should be discussed early in the patient's life.
Collapse
Affiliation(s)
- Zaina Inam
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
7
|
Tanhehco YC, Nathu G, Vasovic LV. Development of curative therapies for sickle cell disease. Front Med (Lausanne) 2022; 9:1055540. [PMID: 36507504 PMCID: PMC9729691 DOI: 10.3389/fmed.2022.1055540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Recent advances in managing Sickle Cell Disease (SCD) significantly improved patient survival and quality of life. Disease-modifying drug therapies such as hydroxyurea, L-glutamine, voxelotor, and crizanlizumab reduce pain crises and severe complications. Allogeneic hematopoietic stem cell transplantation using matched-sibling donors is currently the only standard curative option; however, only a small proportion of patients have such donors. Cord blood and haploidentical transplantation with a modified conditioning regimen have expanded the allogeneic donor pool, making the therapy available to more patients. Gene therapy is a promising cure that is currently undergoing clinical trials and different approaches have demonstrated efficacy. Multidisciplinary expertise is needed in developing the best treatment strategy for patients with SCD.
Collapse
Affiliation(s)
- Yvette C. Tanhehco
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Ghazala Nathu
- Department of Clinical Pathology, Bassett Healthcare Network—Cobleskill Regional Hospital, Cobleskill, NY, United States
| | - Ljiljana V. Vasovic
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
8
|
Is a universal cure for sickle cell disease imminent? Transfus Apher Sci 2022; 61:103532. [DOI: 10.1016/j.transci.2022.103532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
9
|
Shaikh MS, Shahzad Z, Tash EA, Janjua OS, Khan MI, Zafar MS. Human Umbilical Cord Mesenchymal Stem Cells: Current Literature and Role in Periodontal Regeneration. Cells 2022; 11:cells11071168. [PMID: 35406732 PMCID: PMC8997495 DOI: 10.3390/cells11071168] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Periodontal disease can cause irreversible damage to tooth-supporting tissues such as the root cementum, periodontal ligament, and alveolar bone, eventually leading to tooth loss. While standard periodontal treatments are usually helpful in reducing disease progression, they cannot repair or replace lost periodontal tissue. Periodontal regeneration has been demonstrated to be beneficial in treating intraosseous and furcation defects to varied degrees. Cell-based treatment for periodontal regeneration will become more efficient and predictable as tissue engineering and progenitor cell biology advance, surpassing the limitations of present therapeutic techniques. Stem cells are undifferentiated cells with the ability to self-renew and differentiate into several cell types when stimulated. Mesenchymal stem cells (MSCs) have been tested for periodontal regeneration in vitro and in humans, with promising results. Human umbilical cord mesenchymal stem cells (UC-MSCs) possess a great regenerative and therapeutic potential. Their added benefits comprise ease of collection, endless source of stem cells, less immunorejection, and affordability. Further, their collection does not include the concerns associated with human embryonic stem cells. The purpose of this review is to address the most recent findings about periodontal regenerative mechanisms, different stem cells accessible for periodontal regeneration, and UC-MSCs and their involvement in periodontal regeneration.
Collapse
Affiliation(s)
- Muhammad Saad Shaikh
- Department of Oral Biology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi 75510, Pakistan;
| | - Zara Shahzad
- Lahore Medical and Dental College, University of Health Sciences, Lahore 53400, Pakistan;
| | - Esraa Abdulgader Tash
- Department of Oral and Clinical Basic Science, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia;
| | - Omer Sefvan Janjua
- Department of Maxillofacial Surgery, PMC Dental Institute, Faisalabad Medical University, Faisalabad 38000, Pakistan;
| | | | - Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
- Correspondence: ; Tel.: +966-507544691
| |
Collapse
|
10
|
Oudin Doglioni D, Pham-Hung D’Alexandry D’Orengiani AL, Galactéros F, Gay MC. Psychometric characteristics of the Revised Illness Perception Questionnaire (IPQ-R) in adults with sickle cell disease. Health Psychol Behav Med 2021; 10:60-80. [PMID: 34993006 PMCID: PMC8725854 DOI: 10.1080/21642850.2021.2016411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/04/2021] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Sickle cell disease (SCD) is the most frequent monogenic disease worldwide. Psychological and behavioural factors are often reported as playing a significant role in predicting SCD health outcomes. When focusing on adaptation to a specific health condition and its treatment, the Common Sense Model of Health and Illness (CSM) has proven to be of heuristic value. In other health conditions, illness outcomes are directly influenced by illness perception. Therefore, the aim of this study is to explore the psychometric proprieties of the Revised Illness Perception Questionnaire (IPQ-R). DESIGN We performed a cross-sectional assessment on 517 adult patients with sickle cell disease and collected the results of 406 IPQ-R. With these data, we verified the factor structure of the Belief scale and proposed modifications to improve its fit to the data with a confirmatory factor analysis. In addition, we explored the factorial structure of the Causal attribution scale with an exploratory factor analysis. RESULTS The initial model showed poor fit with the data. After structural modifications, elimination of two items with a low loading (model 2), covariance added between items (model 3) and items reallocation (model 4), the last model proposed presented a correct fit with the data. Before doing this model specification, we reviewed and compiled the nine studies that explored the psychometric properties of the IPQ-R in order to highlight all the modifications made by the other authors who have adapted the IPQ-R to a specific population and to allow a comparison with our own modifications. CONCLUSION Considering previous findings, this research suggests further work is needed on the structure of the dimensions of the IPQ-R.
Collapse
Affiliation(s)
- Damien Oudin Doglioni
- Institut Pasteur, Emerging diseases Unit (Global Health Department), Paris, France
- Paris Nanterre University, EA4430 EvaCliPsy/ED139, Nanterre, France
| | | | - Frédéric Galactéros
- Teaching Hospital Henri Mondor, Red blood cell Genetic Diseases Unit (UMGGR), Créteil, France
- French National Referral Centre for sickle cell disease (MCGRE), Créteil, France
| | - Marie-Claire Gay
- Paris Nanterre University, EA4430 EvaCliPsy/ED139, Nanterre, France
| |
Collapse
|
11
|
Kanter J. Gene therapy for sickle cell disease: where we are now? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:174-180. [PMID: 34889358 PMCID: PMC8791177 DOI: 10.1182/hematology.2021000250] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The landscape of sickle cell disease (SCD) treatment continues to evolve rapidly, with new disease-modifying therapies in development and potentially curative options on the horizon. Until recently, allogeneic stem cell transplant has been the only proven cure for SCD. Gene therapy is rising to the forefront of the discussion as a potentially curative or highly disease- modifying option for abating the complications of the disease. Understanding the different types of gene therapy in use, the differences in their end points, and their potential risks and benefits will be key to optimizing the long-term use of this therapy.
Collapse
Affiliation(s)
- Julie Kanter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW This review summarizes the significant biophysical and rheological aspects of red blood cell physiology and pathophysiology in relation to recent advances in microfluidic biomarker assays and emerging targeted or curative intent therapies. RECENT FINDINGS Alterations in red cell biophysical properties and blood rheology have been associated with numerous hematologic and circulatory disorders. Recent advances in biomarker assays enable effective assessment of these biophysical and rheological properties in normoxia or physiological hypoxia in a clinically meaningful way. There are emerging targeted or curative therapies that aim to improve red cell pathophysiology, especially in the context of inherited hemoglobin disorders, such as sickle cell disease. SUMMARY Red cell pathophysiology can be therapeutically targeted and the improvements in membrane and cellular biophysics and blood rheology can now be feasibly assessed via new microfluidic biomarker assays. Recent advances provide a new hope and novel treatment options for major red cell ailments, including inherited hemoglobin disorders, membrane disorders, and other pathologies of the red cell, such as malaria.
Collapse
Affiliation(s)
- Umut A. Gurkan
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH 44106, USA
- Biomedical Engineering Department, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
13
|
Abstract
Beta hemoglobinopathies such as sickle cell disease (SCD) and β-thalassemia (BT) are the most common monogenic diseases worldwide. Both diseases are associated with significant morbidity and mortality. Because patients require lifelong follow-up and care, it also poses a serious burden in health services. Blood transfusions and/or drug therapy ameliorate the signs and symptoms of the disorders but are not curative. Allogeneic hematopoietic cell transplantation (HCT) is currently the only cure but it has several limitations including the paucity of human leukocyte antigen-matched related donors and a high risk of adverse events. Recent advances in hematopoietic stem cell based-gene therapy has made autologous HCT (auto-HCT) a reality. Clinical trials are underway using different gene transfer vectors and cassettes. Data obtained so far with a short-term follow-up has been very encouraging. Patients with SCD engrafted, had sustained production of the transgene and a decreased number of vaso-occlusive crises. Patients with BT were able to decrease the amount of transfusions required or stop transfusions all together. Adverse events observed were mostly associated with the myeloablative conditioning regimen. Long term data on gene persistence and toxicities are still needed. This review focuses on the current state of auto-HCT with gene therapy for SCD and BT. Current clinical trials and their outcome results are summarized.
Collapse
Affiliation(s)
- Yvette C Tanhehco
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Division of Transfusion Medicine, 622 W. 168thStreet, Harkness Pavilion 4-418A, New York, NY, 10032, United States.
| |
Collapse
|
14
|
Rajendiran S, Boyer SW, Forsberg EC. A quantitative hematopoietic stem cell reconstitution protocol: Accounting for recipient variability, tissue distribution and cell half-lives. Stem Cell Res 2020; 50:102145. [PMID: 33486300 PMCID: PMC8239062 DOI: 10.1016/j.scr.2020.102145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/03/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem and progenitor cell (HSPC) transplantation is the paradigm for stem cell therapies. The protocol described here enables quantitative assessment of the body-wide HSPC reconstitution of different mature hematopoietic cells in mice based on their presence in circulating blood. The method determines donor-derived mature cell populations per mouse, over time, by quantitatively obtaining their absolute numbers in the peripheral blood and utilizing previously assessed tissue-distribution factors. A Markov-based birth/death computational model accounts for the drastic differences in mature cell half-lives. By quantifying the number of cells produced and eliminating host variability, the protocol can be used to directly compare the lineage output of different types of HSPCs on a per cell basis, thereby clarifying the lineage potential and expansion capacity of different cell populations. These protocols were developed for hematopoiesis, but can readily be extended to other contexts by simply replacing the cell types and distributions.
Collapse
Affiliation(s)
- Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
15
|
Abstract
Red blood cell (RBC) transfusion is critical in managing acute and chronic complications of sickle cell disease. Alloimmunization and iron overload remain significant complications of transfusion therapy and are minimized with prophylactic Rh and K antigen RBC matching and iron chelation. Matched sibling donor hematopoietic stem cell transplant (HSCT) is a curative therapeutic option. Autologous hematopoietic stem cell (HSC)-based gene therapy has recently shown great promise, for which obtaining sufficient HSCs is essential for success. This article discusses RBC transfusion indications and complications, transfusion support during HSCT, and HSC mobilization and collection for autologous HSCT with gene therapy.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pathology, St. Jude Children's Research Hospital, MS 342, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stella T Chou
- Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, 3615 Civic Center Boulevard, Abramson Research Center Room 316D, Philadelphia, PA 19010, USA.
| |
Collapse
|
16
|
Alatyyat SM, Alasmari HM, Aleid OA, Abdel-Maksoud MS, Elsherbiny N. Umbilical cord stem cells: Background, processing and applications. Tissue Cell 2020; 65:101351. [PMID: 32746993 DOI: 10.1016/j.tice.2020.101351] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/15/2020] [Accepted: 03/15/2020] [Indexed: 12/26/2022]
Abstract
Stem cells have currently gained attention in the field of medicine not only due to their ability to repair dysfunctional or damaged cells, but also they could be used as drug delivery system after being engineered to do so. Human umbilical cord is attractive source for autologous and allogenic stem cells that are currently amenable to treatment of various diseases. Human umbilical cord stem cells are -in contrast to embryonic and fetal stem cells- ethically noncontroversial, inexpensive and readily available source of cells. Umbilical cord, umbilical cord vein, amnion/placenta and Wharton's jelly are all rich of many types of multipotent stem cell populations capable of forming many different cell types. This review will focus on umbilical cord stem cells processing and current application in medicine.
Collapse
Affiliation(s)
- Shumukh M Alatyyat
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Houton M Alasmari
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Omamah A Aleid
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohamed S Abdel-Maksoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|