1
|
Duan Y, Wu C, Lai Z, Yuan Q, Hu N, Liu S, Liu Z. Autonomous Epinephrine Release by KCNJ5 Mutation Drives Familial Thoracic Aortic Aneurysm and Dissection. Hypertension 2025; 82:752-764. [PMID: 39907018 DOI: 10.1161/hypertensionaha.124.23795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The pathophysiology of familial thoracic aortic aneurysm and dissection (TAAD) is linked to genetic variants that affect aortic components. Although hypertension is a risk factor for TAAD, the precise genetic link remains unclear. METHODS A family with autosomal dominant TAAD complicated by hypertension was studied to identify candidate mutations. The effect of the identified mutation on TAAD development was investigated using a clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-generated knock-in mouse model to elucidate the mechanism underlying hypertension-induced TAAD. RESULTS The KCNJ5 p.R242Q mutation was identified in the family and met the criteria for cosegregation, rarity, and conservation. Utilizing our mouse model, we observed that a significant proportion of heterozygous mice with the mutation displayed dilated thoracic aortas. The mutation's allele dose was positively correlated with TAAD incidence following β-aminopropionitrile monofumarate treatment. Pathological changes in the thoracic aorta, including collagen deposition and dilation, elevated transforming growth factor-β activity, and extracellular matrix remodeling, were associated with hypertension. Furthermore, the mutation was found to induce lifelong isolated systolic hypertension, attributable to autonomous epinephrine secretion from the adrenal medulla. Unlike wild-type, mutated KCNJ5 was highly expressed in the adrenal medulla instead of the adrenal cortex. Treatment with the adrenergic β-receptor blocker propranolol reduced systolic hypertension and mitigated TAAD in the heterozygous mice. CONCLUSIONS Familial TAAD may stem from KCNJ5 dysfunction in the G-protein-coupling domain, causing isolated systolic hypertension via increased epinephrine secretion and disruption of thoracic aortic homeostasis. These findings establish a genetic link between systolic hypertension and TAAD.
Collapse
Affiliation(s)
- Yanyu Duan
- Engineering Research Center of Intelligent Acoustic Signals of Jiangxi Province, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (Y.D., C.W., N.H., Z. Liu), Gannan Medical University, Ganzhou, China
- Ganzhou Cardiovascular Rare Disease Diagnosis and Treatment Technology Innovation Center (Y.D., Z. Liu), Gannan Medical University, Ganzhou, China
- Heart Medical Centre (Y.D., Z. Lai, Q.Y., Z. Liu), First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Chenglong Wu
- Engineering Research Center of Intelligent Acoustic Signals of Jiangxi Province, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (Y.D., C.W., N.H., Z. Liu), Gannan Medical University, Ganzhou, China
| | - Zhenghong Lai
- Heart Medical Centre (Y.D., Z. Lai, Q.Y., Z. Liu), First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qunxing Yuan
- Heart Medical Centre (Y.D., Z. Lai, Q.Y., Z. Liu), First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Naixing Hu
- Engineering Research Center of Intelligent Acoustic Signals of Jiangxi Province, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (Y.D., C.W., N.H., Z. Liu), Gannan Medical University, Ganzhou, China
| | - Shaoqiang Liu
- Radiology Department (S.L.), First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Ziyou Liu
- Engineering Research Center of Intelligent Acoustic Signals of Jiangxi Province, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education (Y.D., C.W., N.H., Z. Liu), Gannan Medical University, Ganzhou, China
- Ganzhou Cardiovascular Rare Disease Diagnosis and Treatment Technology Innovation Center (Y.D., Z. Liu), Gannan Medical University, Ganzhou, China
- Heart Medical Centre (Y.D., Z. Lai, Q.Y., Z. Liu), First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
2
|
Jeunemaitre X, Mousseaux E, Frank M, Adham S, Pitocco F, Billon C, Ben Yakhlef M, El Hachmi M, Bura-Rivière A, Lapébie FX, Le Hello C, Laneelle D, Seinturier C, Dieterich K, Lambert M, Dupuis-Girod S, Zuily S, Bal-Theoleyre L, Boulon C, Henneton P, Lu E, Denarié N, Boutouyrie P, Mirault T, Chatellier G, Azizi M. Efficacy of Irbesartan in Celiprolol-Treated Patients With Vascular Ehlers-Danlos Syndrome. Circulation 2025; 151:686-695. [PMID: 39906986 DOI: 10.1161/circulationaha.124.072849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Vascular Ehlers-Danlos syndrome is a rare genetic disorder characterized by defective type III collagen and a high risk of arterial morbidity and mortality. Several cardiovascular drugs are used for treatment, including celiprolol, but no controlled trial in this condition has been conducted to date. We hypothesized the benefit of the addition of an angiotensin II receptor blocker. METHODS A multicenter, randomized, placebo-controlled trial was conducted to assess the efficacy and safety of the angiotensin II receptor blocker irbesartan in adults with vascular Ehlers-Danlos syndrome on stable background celiprolol therapy. Patients were randomized 1:1 to receive irbesartan (150 mg/day titrated to 300 mg/day) or placebo for 2 years. The composite primary outcome was defined as any vascular Ehlers-Danlos syndrome-related fatal or nonfatal arterial event or any new or worsening arterial lesions detected by systematic head-to-pelvis computed tomography angiography or peripheral arterial duplex ultrasound at different time points, using a time-to-first-event analysis. RESULTS Twenty-nine participants (62% female; 40.3±11.3 years of age) were randomized to irbesartan, and 28 (64% female; 40.7±11.0 years of age) were randomized to placebo. The composite primary outcome occurred in 8 of 29 patients (27.6%) receiving irbesartan versus 15 of 28 patients (53.6%) receiving placebo (hazard ratio, 0.42 [95% CI, 0.17, 0.99]; P<0.05). The risk of recurrent symptomatic or nonsymptomatic arterial events was lower with irbesartan than with placebo (risk ratio, 0.37 [95% CI, 0.19, 0.68]; P=0.002). A reduction of progression of arterial lesions was observed at all sites. Irbesartan significantly reduced systolic blood pressure compared with placebo (baseline-adjusted difference of 5.4 mm Hg [P<0.001]), but no relation was observed with the reduction of the primary composite outcome. Eleven episodes of irbesartan-related hypotension were recorded, leading to a downtitration in 4 patients. CONCLUSIONS Compared with placebo, irbesartan reduced the risk of severe symptomatic and asymptomatic arterial events in patients with vascular Ehlers-Danlos syndrome on background celiprolol therapy. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02597361.
Collapse
Affiliation(s)
- Xavier Jeunemaitre
- PARCC (X.J., E.M., P.B., T.M., G.C.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Génomique des Maladies Rares et DMU BioPhyGen (X.J., C. Billon, M.B.Y.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Elie Mousseaux
- PARCC (X.J., E.M., P.B., T.M., G.C.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Radiologie Vasculaire (E.M., F.P.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Michael Frank
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Vasculaire et Centre de Référence des Maladies Artérielles Rares (M.F., S.A., M.E.H., N.D., T.M.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Salma Adham
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Vasculaire et Centre de Référence des Maladies Artérielles Rares (M.F., S.A., M.E.H., N.D., T.M.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
- Service de Médecine Vasculaire, Hôpital Saint-Eloi, CHU Montpellier, France (S.A.)
| | - Francesca Pitocco
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Radiologie Vasculaire (E.M., F.P.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Clarisse Billon
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Génomique des Maladies Rares et DMU BioPhyGen (X.J., C. Billon, M.B.Y.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Molka Ben Yakhlef
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Génomique des Maladies Rares et DMU BioPhyGen (X.J., C. Billon, M.B.Y.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Mohamed El Hachmi
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Vasculaire et Centre de Référence des Maladies Artérielles Rares (M.F., S.A., M.E.H., N.D., T.M.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
- Dipartimento Medicina Molecolare, University La Sapienza, Italy (M.E.H.)
| | - Alessandra Bura-Rivière
- Service de Médecine Vasculaire, CHU de Toulouse, Hôpital Rangueil, Toulouse, France (A.B.-R., F.-X.L.)
- Unité Mixte de Recherche 1295 INSERM, Centre d'Epidémiologie et de Recherche en Santé des Populations, Toulouse III-Paul Sabatier University, Toulouse, France (A.B.-R., F.-X.L.)
| | - François-Xavier Lapébie
- Service de Médecine Vasculaire, CHU de Toulouse, Hôpital Rangueil, Toulouse, France (A.B.-R., F.-X.L.)
- Unité Mixte de Recherche 1295 INSERM, Centre d'Epidémiologie et de Recherche en Santé des Populations, Toulouse III-Paul Sabatier University, Toulouse, France (A.B.-R., F.-X.L.)
| | - Claire Le Hello
- Departement de Médecine Vasculaire, CHU Caen Normandie, Caen, France (C.L.H.)
- Service de Médecine Vasculaire, CHU St-Etienne, Hôpital Nord, St-Etienne, France (C.L.H.)
- INSERM, U1059, Université Jean Monnet, St-Etienne, France (C.L.H.)
| | - Damien Laneelle
- INSERM, COMETE, GIP Cyceron, Université de Caen Normandie, Caen, France (D.L.)
| | | | - Klaus Dieterich
- Service de Génétique Médicale, CHU Grenoble, Université Grenoble Alpes, INSERM 1209, Institut pour l'Avancée des Biosciences, Grenoble University Hospital, France (K.D.)
| | - Marc Lambert
- Unité Médico Chirurgicale Vasculaire, Service de Médecine Interne, Hôpital Claude Huriez, CHRU Lille, France (M.L.)
| | - Sophie Dupuis-Girod
- National HHT Reference Center and Genetics Department, Hospices Civils de Lyon, Femme-Mère-Enfants Hospital, Bron, France (S.D.-G.)
- INSERM, Biosanté Unit U1292, Université Grenoble Alpes, CEA, Grenoble, France (S.D.-G., P.H.)
| | - Stéphane Zuily
- CHRU Nancy, Service de Médecine Vasculaire, Centre de Référence pour les Maladies Auto-immunes et Auto-inflammatoires, Centre de Compétence pour les Maladies Vasculaires Rares, Hôpitaux de Brabois, Vandoeuvre-lès-Nancy, France (S.Z.)
- INSERM, U1116, Défaillance Cardiovasculaire Aiguë et Chronique, Université de Lorraine, Nancy, France (S.Z.)
| | - Laurence Bal-Theoleyre
- AP-HM, CHU La Timone, Centre de Référence Pour le Syndrome de Marfan et Apparentés, Centre Aorte Timone, Université Aix-Marseille, Marseille, France (L.B.-T.)
| | - Carine Boulon
- Hôpital Saint-André, CHU Bordeaux, France (C. Boulon)
| | - Pierrick Henneton
- INSERM, Biosanté Unit U1292, Université Grenoble Alpes, CEA, Grenoble, France (S.D.-G., P.H.)
| | - Estelle Lu
- Unité de Recherche Clinique (E.L., G.C.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Nicolas Denarié
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Vasculaire et Centre de Référence des Maladies Artérielles Rares (M.F., S.A., M.E.H., N.D., T.M.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Pierre Boutouyrie
- PARCC (X.J., E.M., P.B., T.M., G.C.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- Service de Pharmacologie (P.B.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Tristan Mirault
- PARCC (X.J., E.M., P.B., T.M., G.C.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- AP-HP, Hôpital Européen Georges-Pompidou, Service de Médecine Vasculaire et Centre de Référence des Maladies Artérielles Rares (M.F., S.A., M.E.H., N.D., T.M.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Gilles Chatellier
- PARCC (X.J., E.M., P.B., T.M., G.C.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- Unité de Recherche Clinique (E.L., G.C.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Michel Azizi
- CIC1418 (M.A.), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, France
- and Hypertension Department and DMU CARTE (M.A.), AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| |
Collapse
|
3
|
He Z, IJpma AS, Vreeken D, Heijsman D, Rosier K, Verhagen HJM, de Bruin JL, Brüggenwirth HT, Roos-Hesselink JW, Bekkers JA, Huylebroeck DFE, van Beusekom HMM, Creemers JWM, Majoor-Krakauer D. The proprotein convertase FURIN is a novel aneurysm predisposition gene impairing TGF-β signalling. Cardiovasc Res 2024; 120:2278-2292. [PMID: 38636100 PMCID: PMC11687399 DOI: 10.1093/cvr/cvae078] [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: 05/31/2023] [Revised: 01/18/2024] [Accepted: 02/25/2024] [Indexed: 04/20/2024] Open
Abstract
AIMS Aortic aneurysms (AA) frequently involve dysregulation of transforming growth factor β (TGF-β)-signalling in the aorta. Here, FURIN was tested as aneurysm predisposition gene given its role as proprotein convertase in pro-TGF-β maturation. METHODS AND RESULTS Rare FURIN variants were detected by whole-exome sequencing of 781 unrelated aortic aneurysm patients and affected relatives. Thirteen rare heterozygous FURIN variants occurred in 3.7% (29) unrelated index AA patients, of which 72% had multiple aneurysms or a dissection. FURIN maturation and activity of these variants were decreased in vitro. Patient-derived fibroblasts showed decreased pro-TGF-β processing, phosphorylation of downstream effector SMAD2 and kinases ERK1/2, and steady-state mRNA levels of the TGF-β-responsive ACTA2 gene. In aortic tissue, collagen and fibrillin fibres were affected. One variant (R745Q), observed in 10 unrelated cases, affected TGF-β signalling variably, indicating effect modification by individual genetic backgrounds. CONCLUSION FURIN is a novel, frequent genetic predisposition for abdominal-, thoracic-, and multiple aortic or middle sized artery aneurysms in older patients, by affecting intracellular TGF-β signalling, depending on individual genetic backgrounds.
Collapse
Affiliation(s)
- Zongsheng He
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Arne S IJpma
- Department of Pathology, Erasmus MC University Medical Center, Dr. Molewater 40, PO BOX 2040, Rotterdam 3000 CA, The Netherlands
| | - Dianne Vreeken
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - Daphne Heijsman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| | - Karen Rosier
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Hence J M Verhagen
- Department of Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Jorg L de Bruin
- Department of Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Hennie T Brüggenwirth
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| | - Jolien W Roos-Hesselink
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - Jos A Bekkers
- Department of Cardiothoracic Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Danny F E Huylebroeck
- Department of Cell Biology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Heleen M M van Beusekom
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - John W M Creemers
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Danielle Majoor-Krakauer
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| |
Collapse
|
4
|
García-Domínguez DJ, López-Enríquez S, Alba G, Garnacho C, Jiménez-Cortegana C, Flores-Campos R, de la Cruz-Merino L, Hajji N, Sánchez-Margalet V, Hontecillas-Prieto L. Cancer Nano-Immunotherapy: The Novel and Promising Weapon to Fight Cancer. Int J Mol Sci 2024; 25:1195. [PMID: 38256268 PMCID: PMC10816838 DOI: 10.3390/ijms25021195] [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: 12/28/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer is a complex disease that, despite advances in treatment and the greater understanding of the tumor biology until today, continues to be a prevalent and lethal disease. Chemotherapy, radiotherapy, and surgery are the conventional treatments, which have increased the survival for cancer patients. However, the complexity of this disease together with the persistent problems due to tumor progression and recurrence, drug resistance, or side effects of therapy make it necessary to explore new strategies that address the challenges to obtain a positive response. One important point is that tumor cells can interact with the microenvironment, promoting proliferation, dissemination, and immune evasion. Therefore, immunotherapy has emerged as a novel therapy based on the modulation of the immune system for combating cancer, as reflected in the promising results both in preclinical studies and clinical trials obtained. In order to enhance the immune response, the combination of immunotherapy with nanoparticles has been conducted, improving the access of immune cells to the tumor, antigen presentation, as well as the induction of persistent immune responses. Therefore, nanomedicine holds an enormous potential to enhance the efficacy of cancer immunotherapy. Here, we review the most recent advances in specific molecular and cellular immunotherapy and in nano-immunotherapy against cancer in the light of the latest published preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
| | - Soledad López-Enríquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Gonzalo Alba
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain;
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Rocío Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Department of Medicine, University of Seville, 41009 Seville, Spain
| | - Nabil Hajji
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Cancer Division, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, 41009 Seville, Spain
| |
Collapse
|
5
|
Seeburun S, Wu S, Hemani D, Pham L, Ju D, Xie Y, Kata P, Li L. Insights into elastic fiber fragmentation: Mechanisms and treatment of aortic aneurysm in Marfan syndrome. Vascul Pharmacol 2023; 153:107215. [PMID: 37640090 PMCID: PMC10872825 DOI: 10.1016/j.vph.2023.107215] [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: 08/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutations in fibrillin 1 (FBN1) gene. These mutations result in defects in the skeletal, ocular, and cardiovascular systems. Aortic aneurysm is the leading cause of premature mortality in untreated MFS patients. Elastic fiber fragmentation in the aortic vessel wall is a hallmark of MFS-associated aortic aneurysms. FBN1 mutations result in FBN1 fragments that also contribute to elastic fiber fragmentation. Although recent research has advanced our understanding of MFS, the contribution of elastic fiber fragmentation to the pathogenesis of aneurysm formation remains poorly understood. This review provides a comprehensive overview of the molecular mechanisms of elastic fiber fragmentation and its role in the pathogenesis of aortic aneurysm progression. Increased comprehension of elastic fragmentation has significant clinical implications for developing targeted interventions to block aneurysm progression, which would benefit not only individuals with Marfan syndrome but also other patients with aneurysms. Moreover, this review highlights an overlooked connection between inhibiting aneurysm and the restoration of elastic fibers in the vessel wall with various aneurysm inhibitors, including drugs and chemicals. Investigating the underlying molecular mechanisms could uncover innovative therapeutic strategies to inhibit elastin fragmentation and prevent the progression of aneurysms.
Collapse
Affiliation(s)
- Sheilabi Seeburun
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Shichao Wu
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Darshi Hemani
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Lucynda Pham
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Donghong Ju
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Youming Xie
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Priyaranjan Kata
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Li Li
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| |
Collapse
|
6
|
Doherty EL, Aw WY, Warren EC, Hockenberry M, Whitworth CP, Krohn G, Howell S, Diekman BO, Legant WR, Nia HT, Hickey AJ, Polacheck WJ. Patient-derived extracellular matrix demonstrates role of COL3A1 in blood vessel mechanics. Acta Biomater 2023; 166:346-359. [PMID: 37187299 PMCID: PMC10330735 DOI: 10.1016/j.actbio.2023.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
Vascular Ehlers-Danlos Syndrome (vEDS) is a rare autosomal dominant disease caused by mutations in the COL3A1 gene, which renders patients susceptible to aneurysm and arterial dissection and rupture. To determine the role of COL3A1 variants in the biochemical and biophysical properties of human arterial ECM, we developed a method for synthesizing ECM directly from vEDS donor fibroblasts. We found that the protein content of the ECM generated from vEDS donor fibroblasts differed significantly from ECM from healthy donors, including upregulation of collagen subtypes and other proteins related to ECM structural integrity. We further found that ECM generated from a donor with a glycine substitution mutation was characterized by increased glycosaminoglycan content and unique viscoelastic mechanical properties, including increased time constant for stress relaxation, resulting in a decrease in migratory speed of human aortic endothelial cells when seeded on the ECM. Collectively, these results demonstrate that vEDS patient-derived fibroblasts harboring COL3A1 mutations synthesize ECM that differs in composition, structure, and mechanical properties from healthy donors. These results further suggest that ECM mechanical properties could serve as a prognostic indicator for patients with vEDS, and the insights provided by the approach demonstrate the broader utility of cell-derived ECM in disease modeling. STATEMENT OF SIGNIFICANCE: The role of collagen III ECM mechanics remains unclear, despite reported roles in diseases including fibrosis and cancer. Here, we generate fibrous, collagen-rich ECM from primary donor cells from patients with vascular Ehlers-Danlos syndrome (vEDS), a disease caused by mutations in the gene that encodes collagen III. We observe that ECM grown from vEDS patients is characterized by unique mechanical signatures, including altered viscoelastic properties. By quantifying the structural, biochemical, and mechanical properties of patient-derived ECM, we identify potential drug targets for vEDS, while defining a role for collagen III in ECM mechanics more broadly. Furthermore, the structure/function relationships of collagen III in ECM assembly and mechanics will inform the design of substrates for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth L Doherty
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily C Warren
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Max Hockenberry
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Chloe P Whitworth
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Grace Krohn
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Stefanie Howell
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Wesley R Legant
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Hadi Tavakoli Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Anthony J Hickey
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Pediatric Heart Network Trial of Losartan vs. Atenolol in Children and Young Adults with Marfan Syndrome: Impact on Prescription Practices. Pediatr Cardiol 2023; 44:618-623. [PMID: 35902413 DOI: 10.1007/s00246-022-02976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
The Pediatric Heart Network (PHN) trial showed similar efficacy of β-blockers (BB) and angiotensin receptor blockers (ARB) for aortic root dilation in Marfan syndrome, but the impact on prescription practices is unknown. We hypothesized BB and ARB prescriptions would increase after the trial results were published (2014). Prescription data (2007-2016) were obtained from outpatient encounters (IBM Marketscan) for Marfan syndrome patients (6 months-25 years old). Excluding 2014 as a washout period, we analyzed two intervals: 2007-2013 and 2015-2016. Medication categories included BB, ARB, angiotensin converting enzyme inhibitors (ACEI), combination (BB/ARB and/or BB/ACEI), and no drug. Interrupted time-series analysis assessed immediate level change after publication and change in slope for the trend pre- and post-publication. Odds ratios (OR) and 95% confidence intervals from logistic regressions and generalized estimating equation methods accounted for correlation of prescriptions within patients. In 1499 patients (age 14.1 ± 6.1 years, 59% female) seen 2007-2013, BB trended lower [OR 0.91 (0.89, 0.93), p < 0.001] and ARB trended higher [OR 1.12 (1.07, 1.18), p < 0.001], while combination, ACEI, and no drug remained stable. This trend persisted, but was not significant, for BB [OR 0.54 (0.27, 1.08), p = 0.37] and ARB [OR 1.91 (0.55, 6.69), p = 0.31] in 2015-2016. Combination, ACEI, and no drug remained similar. In short term follow-up, changes in prescription practices following publication of the PHN trial were not statistically significant. This may be due to a change seen prior to publication with early adoption of ARBs that was maintained after confirmation of their effectiveness.
Collapse
|
8
|
Taghipour YD, Zarebkohan A, Salehi R, Rahimi F, Torchilin VP, Hamblin MR, Seifalian A. An update on dual targeting strategy for cancer treatment. J Control Release 2022; 349:67-96. [PMID: 35779656 DOI: 10.1016/j.jconrel.2022.06.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/18/2022]
Abstract
The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.
Collapse
Affiliation(s)
- Yasamin Davatgaran Taghipour
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine and Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, South Africa
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
9
|
Făgărășan A, Săsăran MO. The Predictive Role of Plasma Biomarkers in the Evolution of Aortopathies Associated with Congenital Heart Malformations. Int J Mol Sci 2022; 23:ijms23094993. [PMID: 35563383 PMCID: PMC9102091 DOI: 10.3390/ijms23094993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Dilatation of the aorta is a constantly evolving condition that can lead to the ultimate life-threatening event, acute aortic dissection. Recent research has tried to identify quantifiable biomarkers, with both diagnostic and prognostic roles in different aortopathies. Most studies have focused on the bicuspid aortic valve, the most frequent congenital heart disease (CHD), and majorly evolved around matrix metalloproteinases (MMPs). Other candidate biomarkers, such as asymmetric dimethylarginine, soluble receptor for advanced glycation end-products or transforming growth factor beta have also gained a lot of attention recently. Most of the aortic anomalies and dilatation-related studies have reported expression variation of tissular biomarkers. The ultimate goal remains, though, the identification of biomarkers among the serum plasma, with the upregulation of circulating MMP-1, MMP-2, MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), asymmetric dimethylarginine (ADMA), soluble receptor for advanced glycation end-products (sRAGE) and transforming growth factor beta (TGF-β) being reported in association to several aortopathies and related complications in recent research. These molecules are apparently quantifiable from the early ages and have been linked to several CHDs and hereditary aortopathies. Pediatric data on the matter is still limited, and further studies are warranted to elucidate the role of plasmatic biomarkers in the long term follow-up of potentially evolving congenital aortopathies.
Collapse
Affiliation(s)
- Amalia Făgărășan
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Târgu Mureș, Romania;
| | - Maria Oana Săsăran
- Department of Pediatrics III, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Târgu Mureș, Romania
- Correspondence: ; Tel.: +40-720-332-503
| |
Collapse
|
10
|
Amino Acid and Phospholipid Metabolism as an Indicator of Inflammation and Subtle Cardiomyopathy in Patients with Marfan Syndrome. Metabolites 2021; 11:metabo11120805. [PMID: 34940564 PMCID: PMC8707072 DOI: 10.3390/metabo11120805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with Marfan syndrome (MFS) have an increased risk of aortic aneurysm formation, dissection and development of a subtle cardiomyopathy. We analyzed amino acid and lipid metabolic pathways in MFS patients, seeking biomarker patterns as potential monitoring tools of cardiovascular risk with deterioration of myocardial function. We assessed myocardial function in 24 adult MFS patients and compared traditional laboratory values and mass spectrometry-based amino acid, phospholipid and acylcarnitine metabolomes in patients with those in healthy controls. Analytes for which values differed between patients and controls were subjected to regression analysis. A high proportion of patients had signs of impaired diastolic function and elevated serum levels of NT-proBNP. Patients had lower serum levels of taurine, histidine and PCaeC42:3 than controls. The evidence of diastolic dysfunction, aortic root dimensions and history of aortic root surgery correlated with NT-proBNP and taurine levels. Alterations in serum levels of metabolism derived analytes link MFS pathophysiology with inflammation, oxidative stress and incipient cardiomyopathy.
Collapse
|
11
|
Toral M, de la Fuente-Alonso A, Campanero MR, Redondo JM. The NO signalling pathway in aortic aneurysm and dissection. Br J Pharmacol 2021; 179:1287-1303. [PMID: 34599830 DOI: 10.1111/bph.15694] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Recent studies have shown that NO is a central mediator in diseases associated with thoracic aortic aneurysm, such as Marfan syndrome. The progressive dilation of the aorta in thoracic aortic aneurysm ultimately leads to aortic dissection. Unfortunately, current medical treatments have neither halt aortic enlargement nor prevented rupture, leaving surgical repair as the only effective treatment. There is therefore a pressing need for effective therapies to delay or even avoid the need for surgical repair in thoracic aortic aneurysm patients. Here, we summarize the mechanisms through which NO signalling dysregulation causes thoracic aortic aneurysm, particularly in Marfan syndrome. We discuss recent advances based on the identification of new Marfan syndrome mediators related to pathway overactivation that represent potential disease biomarkers. Likewise, we propose iNOS, sGC and PRKG1, whose pharmacological inhibition reverses aortopathy in Marfan syndrome mice, as targets for therapeutic intervention in thoracic aortic aneurysm and are candidates for clinical trials.
Collapse
Affiliation(s)
- Marta Toral
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andrea de la Fuente-Alonso
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel R Campanero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
12
|
Arce C, Rodríguez-Rovira I, De Rycke K, Durán K, Campuzano V, Fabregat I, Jiménez-Altayó F, Berraondo P, Egea G. Anti-TGFβ (Transforming Growth Factor β) Therapy With Betaglycan-Derived P144 Peptide Gene Delivery Prevents the Formation of Aortic Aneurysm in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2021; 41:e440-e452. [PMID: 34162229 DOI: 10.1161/atvbaha.121.316496] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective We investigated the effect of a potent TGFβ (transforming growth factor β) inhibitor peptide (P144) from the betaglycan/TGFβ receptor III on aortic aneurysm development in a Marfan syndrome mouse model. Approach and Results We used a chimeric gene encoding the P144 peptide linked to apolipoprotein A-I via a flexible linker expressed by a hepatotropic adeno-associated vector. Two experimental approaches were performed: (1) a preventive treatment where the vector was injected before the onset of the aortic aneurysm (aged 4 weeks) and followed-up for 4 and 20 weeks and (2) a palliative treatment where the vector was injected once the aneurysm was formed (8 weeks old) and followed-up for 16 weeks. We evaluated the aortic root diameter by echocardiography, the aortic wall architecture and TGFβ signaling downstream effector expression of pSMAD2 and pERK1/2 by immunohistomorphometry, and Tgfβ1 and Tgfβ2 mRNA expression levels by real-time polymerase chain reaction. Marfan syndrome mice subjected to the preventive approach showed no aortic dilation in contrast to untreated Marfan syndrome mice, which at the same end point age already presented the aneurysm. In contrast, the palliative treatment with P144 did not halt aneurysm progression. In all cases, P144 improved elastic fiber morphology and normalized pERK1/2-mediated TGFβ signaling. Unlike the palliative treatment, the preventive treatment reduced Tgfβ1 and Tgfβ2 mRNA levels. Conclusions P144 prevents the onset of aortic aneurysm but not its progression. Results indicate the importance of reducing the excess of active TGFβ signaling during the early stages of aortic disease progression.
Collapse
Affiliation(s)
- Cristina Arce
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain (C.A., I.R.-R., K.D.R., V.C., G.E.)
| | - Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain (C.A., I.R.-R., K.D.R., V.C., G.E.)
| | - Karo De Rycke
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain (C.A., I.R.-R., K.D.R., V.C., G.E.)
| | - Karina Durán
- Department of Cardiology, Hospital Clínic y Provincial de Barcelona, Spain (K.D.)
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain (C.A., I.R.-R., K.D.R., V.C., G.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain (V.C.)
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL) and Centro de Investigación Biomédica en Red de Enfermedades Hepático-Digestivas (CIBEREHD), ISCIII, Spain (I.F.)
| | - Francesc Jiménez-Altayó
- Department of Therapeutic Pharmacology and Toxicology, School of Medicine, Neuroscience Institute, Autonomous University of Barcelona, Bellaterra, Spain (F.J.-A.)
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, CIMA University of Navarra, Pamplona, Spain (P.B.)
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain (P.B.)
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain (C.A., I.R.-R., K.D.R., V.C., G.E.)
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (G.E.)
| |
Collapse
|
13
|
Mirtajaddini M, Khajali Z, Naderi N, Rezaeian N. Does myocardial injury rapidly progress in marfan syndrome following COVID-19? Res Cardiovasc Med 2021. [DOI: 10.4103/rcm.rcm_10_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
14
|
van Dorst DCH, de Wagenaar NP, van der Pluijm I, Roos-Hesselink JW, Essers J, Danser AHJ. Transforming Growth Factor-β and the Renin-Angiotensin System in Syndromic Thoracic Aortic Aneurysms: Implications for Treatment. Cardiovasc Drugs Ther 2020; 35:1233-1252. [PMID: 33283255 PMCID: PMC8578102 DOI: 10.1007/s10557-020-07116-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Thoracic aortic aneurysms (TAAs) are permanent pathological dilatations of the thoracic aorta, which can lead to life-threatening complications, such as aortic dissection and rupture. TAAs frequently occur in a syndromic form in individuals with an underlying genetic predisposition, such as Marfan syndrome (MFS) and Loeys-Dietz syndrome (LDS). Increasing evidence supports an important role for transforming growth factor-β (TGF-β) and the renin-angiotensin system (RAS) in TAA pathology. Eventually, most patients with syndromic TAAs require surgical intervention, as the ability of present medical treatment to attenuate aneurysm growth is limited. Therefore, more effective medical treatment options are urgently needed. Numerous clinical trials investigated the therapeutic potential of angiotensin receptor blockers (ARBs) and β-blockers in patients suffering from syndromic TAAs. This review highlights the contribution of TGF-β signaling, RAS, and impaired mechanosensing abilities of aortic VSMCs in TAA formation. Furthermore, it critically discusses the most recent clinical evidence regarding the possible therapeutic benefit of ARBs and β-blockers in syndromic TAA patients and provides future research perspectives and therapeutic implications.
Collapse
Affiliation(s)
- Daan C H van Dorst
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nathalie P de Wagenaar
- Department of Molecular Genetics, Erasmus University Medical Center, Room Ee702b, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.,Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center, Room Ee702b, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jolien W Roos-Hesselink
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Room Ee702b, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands. .,Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands. .,Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Baderkhan H, Wanhainen A, Stenborg A, Stattin EL, Björck M. Celiprolol Treatment in Patients with Vascular Ehlers-Danlos Syndrome. Eur J Vasc Endovasc Surg 2020; 61:326-331. [PMID: 33223285 DOI: 10.1016/j.ejvs.2020.10.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/17/2020] [Accepted: 10/22/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Vascular Ehlers-Danlos syndrome (vEDS) is a rare monogenetic disease caused by pathogenic variants in procollagen 3A1. Arterial rupture is the most serious clinical manifestation. A randomised controlled trial, the Beta-Blockers in Ehlers-Danlos Syndrome Treatment (BBEST) trial, reported a significant protective effect of the beta blocker celiprolol. The aim was to study the outcome of celiprolol treatment in a cohort of Swedish patients with vEDS. METHODS Uppsala is a national referral centre for patients with vEDS. They are assessed by vascular surgeons, angiologists, and clinical geneticists. Family history, previous and future clinical events, medication, and side effects are registered. Celiprolol was administered twice daily and titrated up to a maximum dose of 400 mg daily. Logistic regression was used to analyse predictors of vascular events. RESULTS Forty patients with pathogenic sequence variants in COL3A1 were offered treatment with celiprolol in the period 2011-2019. The median follow up was 22 months (range 1-98 months); total follow up was 106 patient years. In two patients, uptitration of the dose is ongoing. Of the remaining 38, 26 (65%) patients reached the target dose of 400 mg daily. Dose uptitration was unsuccessful in six patients because of side effects; one died before reaching the maximum dose, and five terminated the treatment. Five major vascular events occurred; four were fatal (ruptured ascending aorta; aortic rupture after type B dissection; ruptured cerebral aneurysm; and ruptured pulmonary artery). One bled from a branch of the internal iliac artery, which was successfully coiled endovascularly. The annual risk of a major vascular event was 4.7% (n = 5/106), similar to the treatment arm of the BBEST trial (5%) and lower than in the control arm of the same trial (12%). No significant predictor of vascular events was identified. CONCLUSION Treatment with celiprolol is tolerated in most patients with vEDS. Despite fatal vascular events, these observations suggest that celiprolol may have a protective effect in vEDS.
Collapse
Affiliation(s)
- Hassan Baderkhan
- Department of Surgical Sciences, Vascular Surgery, Uppsala, Sweden.
| | - Anders Wanhainen
- Department of Surgical Sciences, Vascular Surgery, Uppsala, Sweden
| | | | - Eva-Lena Stattin
- Department of Clinical Genetics, all Uppsala University, Uppsala, Sweden
| | - Martin Björck
- Department of Surgical Sciences, Vascular Surgery, Uppsala, Sweden
| |
Collapse
|
16
|
Aguado T, García M, García A, Ferrer-Mayorga G, Martínez-Santamaría L, del Río M, Botella LM, Sánchez-Puelles JM. Raloxifene and n-Acetylcysteine Ameliorate TGF-Signalling in Fibroblasts from Patients with Recessive Dominant Epidermolysis Bullosa. Cells 2020; 9:E2108. [PMID: 32947957 PMCID: PMC7565802 DOI: 10.3390/cells9092108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a severe skin disease caused by mutation of the COL7A1 gene. RDEB is associated with high levels of TGF-β1, which is likely to be involved in the fibrosis that develops in this disease. Endoglin (CD105) is a type III coreceptor for TGF-β1 and its overexpression in fibroblasts deregulates physiological Smad/Alk1/Alk5 signalling, repressing the synthesis of TGF-β1 and extracellular matrix (ECM) proteins. Raloxifene is a specific estrogen receptor modulator designated as an orphan drug for hereditary hemorrhagic telangiectasia, a rare vascular disease. Raloxifene stimulates endoglin synthesis, which could attenuate fibrosis. By contrast, the antioxidant N-acetylcysteine may have therapeutic value to rectify inflammation, fibrosis and endothelial dysfunction. Thus, we present here a repurposing strategy based on the molecular and functional screening of fibroblasts from RDEB patients with these drugs, leading us to propose the repositioning of these two well-known drugs currently in clinical use, raloxifene and N-acetylcysteine, to counteract fibrosis and inflammation in RDEB. Both compounds modulate the profibrotic events that may ultimately be responsible for the clinical manifestations in RDEB, suggesting that these findings may also be relevant for other diseases in which fibrosis is an important pathophysiological event.
Collapse
Affiliation(s)
- Tania Aguado
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, U-707 CIBERER, 28040 Madrid, Spain;
| | - Marta García
- Departament of Biomedical Engineering, Universidad Carlos III, 28911 Madrid, Spain; (M.G.); (A.G.); (L.M.-S.); (M.d.R.)
- Spanish Network of Research Groups on Rare Diseases (CIBERER) U714, 28911 Madrid, Spain
- Foundation of the Institute for Health Research, Jiménez Díaz Foundation, 28040 Madrid, Spain
| | - Adela García
- Departament of Biomedical Engineering, Universidad Carlos III, 28911 Madrid, Spain; (M.G.); (A.G.); (L.M.-S.); (M.d.R.)
- Spanish Network of Research Groups on Rare Diseases (CIBERER) U714, 28911 Madrid, Spain
- Foundation of the Institute for Health Research, Jiménez Díaz Foundation, 28040 Madrid, Spain
| | - Gemma Ferrer-Mayorga
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Lucía Martínez-Santamaría
- Departament of Biomedical Engineering, Universidad Carlos III, 28911 Madrid, Spain; (M.G.); (A.G.); (L.M.-S.); (M.d.R.)
- Spanish Network of Research Groups on Rare Diseases (CIBERER) U714, 28911 Madrid, Spain
- Foundation of the Institute for Health Research, Jiménez Díaz Foundation, 28040 Madrid, Spain
| | - Marcela del Río
- Departament of Biomedical Engineering, Universidad Carlos III, 28911 Madrid, Spain; (M.G.); (A.G.); (L.M.-S.); (M.d.R.)
- Spanish Network of Research Groups on Rare Diseases (CIBERER) U714, 28911 Madrid, Spain
- Foundation of the Institute for Health Research, Jiménez Díaz Foundation, 28040 Madrid, Spain
| | - Luisa-María Botella
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, U-707 CIBERER, 28040 Madrid, Spain;
| | - José-María Sánchez-Puelles
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, U-707 CIBERER, 28040 Madrid, Spain;
| |
Collapse
|
17
|
Heidarpour M, Taheri M, Akhavan A, Goli P, Kefayat A. Investigation of HER-2 Expression an Its Correlation with Clinicopathological Parameters and Overall Survival of Esophageal Squamous Cell Carcinoma Patients. IRANIAN JOURNAL OF PATHOLOGY 2020; 15:274-281. [PMID: 32944039 PMCID: PMC7477677 DOI: 10.30699/ijp.2020.113829.2235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Background & Objective: Human epidermal growth factor receptor 2 (HER-2) exhibits a vast range of expression in esophageal squamous cell carcinoma (ESCC) patients as a biomarker. This paper aimed to investigate HER-2 expression and clinicopathological parameters of esophageal SCC. Methods: HER-2 expression was assessed in 102 ESCC patients by immunohistochemistry. The HER-2 staining intensity , according to the Gastric HER2 Biomarker1.0.0.1 version of the college of American pathologists (CAP) protocol for gastric and gastroesophageal junction cancers, was graded as 0 (no reactivity in any of the cancer cells’ membranes); 1+ (pale or hardly noticeable reactivity in the membrane of cancer cells’ cluster [≥ 5 neoplastic cells] regardless of the positive cancer cells’ percentage); 2+ (weak-to-moderate complete, basolateral, or lateral membranous reactivity regardless of the positive cancer cells’ percentage); and 3+ ( strong complete, basolateral, or lateral reactivity in the membrane of the cancer cell cluster regardless of the positive cancer cells’ percentage).In this regard, 3+ scored samples were considered as positive. If HER-2 expression was scored 2+, an additional fluorescence in situ hybridization (FISH) was performed. Fisher's exact test was employed for investigating the correlation of HER-2 expression status with patients’ clinicopathological characteristics (including age, gender, tumor location, stage, grade, infiltration level, venous invasion, lymphatic invasion, and tumor recurrence). Kaplan-Meier analysis was done for the patients’ survival assessments. Results: Five patients (~5%) were HER-2 positive and no significant association was observed between HER-2 expression and clinicopathological properties. In addition, HER-2 expression status exhibited no significant association with the patients’ overall survival (P=0.9299). Conclusion: HER-2 is not a suitable prognostic biomarker for Iranian ESCC patients.
Collapse
Affiliation(s)
- Mitra Heidarpour
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehran Taheri
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Akhavan
- Department of Radiation Oncology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Goli
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
18
|
Fang T, Zhang J, Zuo T, Wu G, Xu Y, Yang Y, Yang J, Shen Q. Chemo-Photothermal Combination Cancer Therapy with ROS Scavenging, Extracellular Matrix Depletion, and Tumor Immune Activation by Telmisartan and Diselenide-Paclitaxel Prodrug Loaded Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2020; 12:31292-31308. [PMID: 32551473 DOI: 10.1021/acsami.0c10416] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Extracellular matrix (ECM) accumulating in the tumor microenvironment (TME) is generated by tumor-associated fibroblasts. It can elevate interstitial fluid pressure and form dense barriers in tumor tissues. Consequently, nanocarriers are hindered from permeating into deeper tumor sites. Thus, the programmed drug-releasing nanoparticles, G(TM)PPSP, were developed for TME remodeling and breast cancer therapy. Gelatin nanoparticles were linked with platinum nanoparticles (PtNPs) to obtain G(TM)PPSP with a size of 214.0 ± 5.0 nm. Telmisartan (TM) was loaded in gelatin nanoparticles. Paclitaxel (PTX) was attached to PtNPs via a dual redox responsive diselenide bond. TM release was mediated by MMP-2 because of gelatin degradation in TME, and then intracellular PTX was released because of diselenide linkage fracture triggered by ROS or glutathione. ECM was depleted owing to TGF-β downregulation by TM and direct ablation by the photothermal effect of PtNPs. 4T1 tumor progression was inhibited by PTX chemotherapy, intracellular ROS scavenging of PtNPs, and photothermal therapy (PTT). The tumor spheroid penetration assay proved G(TM)PPSP could permeate into deep tumor regions when MMP-2 existed. In vivo antitumor experiments implied G(TM)PPSP with PTT could inhibit tumor growth effectively and remodel TME via ECM depletion and immunity activation, indicating the potential of G(TM)PPSP-based chemo-photothermal combination therapy for breast cancer treatment.
Collapse
Affiliation(s)
- Tianxu Fang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Tiantian Zuo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Guangyu Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200120, P. R. China
| | - Yingxin Xu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Yifan Yang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jie Yang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Qi Shen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
19
|
Zhou Y, Chen X, Cao J, Gao H. Overcoming the biological barriers in the tumor microenvironment for improving drug delivery and efficacy. J Mater Chem B 2020; 8:6765-6781. [PMID: 32315375 DOI: 10.1039/d0tb00649a] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The delivery of drugs to tumors by nanoparticles is a rapidly growing field. However, the complex tumor microenvironment (TME) barriers greatly hinder drug delivery to tumors. In this study, we first summarized the barriers in TME, including anomalous vasculature, rigid extracellular matrix, hypoxia, acidic pH, irregular enzyme level, altered metabolism pathway and immunosuppressive conditions. To overcome these barriers, many strategies have been developed, such as modulating TME, active targeting by ligand modification and biomimetic strategies, and TME-responsive drug delivery strategies to improve nanoparticle penetration, cellular uptake and drug release. Although extensive progress has been achieved, there are still many challenges, which are discussed in the last section. Overall, we carefully discuss the landscape of TME, development for improving drug delivery, and challenges that need to be further addressed.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| | | | | | | |
Collapse
|
20
|
Garcia-Puig A, Mosquera JL, Jiménez-Delgado S, García-Pastor C, Jorba I, Navajas D, Canals F, Raya A. Proteomics Analysis of Extracellular Matrix Remodeling During Zebrafish Heart Regeneration. Mol Cell Proteomics 2019; 18:1745-1755. [PMID: 31221719 PMCID: PMC6731076 DOI: 10.1074/mcp.ra118.001193] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Adult zebrafish, in contrast to mammals, are able to regenerate their hearts in response to injury or experimental amputation. Our understanding of the cellular and molecular bases that underlie this process, although fragmentary, has increased significantly over the last years. However, the role of the extracellular matrix (ECM) during zebrafish heart regeneration has been comparatively rarely explored. Here, we set out to characterize the ECM protein composition in adult zebrafish hearts, and whether it changed during the regenerative response. For this purpose, we first established a decellularization protocol of adult zebrafish ventricles that significantly enriched the yield of ECM proteins. We then performed proteomic analyses of decellularized control hearts and at different times of regeneration. Our results show a dynamic change in ECM protein composition, most evident at the earliest (7 days postamputation) time point analyzed. Regeneration associated with sharp increases in specific ECM proteins, and with an overall decrease in collagens and cytoskeletal proteins. We finally tested by atomic force microscopy that the changes in ECM composition translated to decreased ECM stiffness. Our cumulative results identify changes in the protein composition and mechanical properties of the zebrafish heart ECM during regeneration.
Collapse
Affiliation(s)
- Anna Garcia-Puig
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain; §Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Jose Luis Mosquera
- ¶Bioinformatics Unit, Institut d'Investigació Biomèdica de Bellvitge IDIBELL), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Senda Jiménez-Delgado
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Cristina García-Pastor
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain
| | - Ignasi Jorba
- ‖Institute for Bioengineering of Catalonia (IBEC), Barcelona Science Park, Baldiri Reixac 15-21, 08028 Barcelona, Spain; **Unit of Biophysics and Bioengineering, Department of Physiological Sciences I, School of Medicine, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; ‡‡Center for Networked Biomedical Research on Respiratory Diseases (CIBERES), 08036 Barcelona, Spain
| | - Daniel Navajas
- ‖Institute for Bioengineering of Catalonia (IBEC), Barcelona Science Park, Baldiri Reixac 15-21, 08028 Barcelona, Spain; **Unit of Biophysics and Bioengineering, Department of Physiological Sciences I, School of Medicine, University of Barcelona, Casanova 143, 08036 Barcelona, Spain; ‡‡Center for Networked Biomedical Research on Respiratory Diseases (CIBERES), 08036 Barcelona, Spain
| | - Francesc Canals
- §§Proteomics group, Vall d'Hebron Institut of Oncology (VHIO), Cellex center, Natzaret 115-117, 08035 Barcelona, Spain
| | - Angel Raya
- ‡Center of Regenerative Medicine in Barcelona (CMRB), 3rd Floor Hospital Duran i Reynals, Avinguda de la Gran Via 199-203, 08908 Hospitalet de Llobregat (Barcelona), Spain; §Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08908 Hospitalet de Llobregat (Barcelona), Spain; ¶¶Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
21
|
Emrich F, Penov K, Arakawa M, Dhablania N, Burdon G, Pedroza AJ, Koyano TK, Kim YM, Raaz U, Connolly AJ, Iosef C, Fischbein MP. Anatomically specific reactive oxygen species production participates in Marfan syndrome aneurysm formation. J Cell Mol Med 2019; 23:7000-7009. [PMID: 31402541 PMCID: PMC6787454 DOI: 10.1111/jcmm.14587] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Marfan syndrome (MFS) is a connective tissue disorder that results in aortic root aneurysm formation. Reactive oxygen species (ROS) seem to play a role in aortic wall remodelling in MFS, although the mechanism remains unknown. MFS Fbn1C1039G/+ mouse root/ascending (AS) and descending (DES) aortic samples were examined using DHE staining, lucigenin‐enhanced chemiluminescence (LGCL), Verhoeff's elastin‐Van Gieson staining (elastin breakdown) and in situ zymography for protease activity. Fbn1C1039G/+ AS‐ or DES‐derived smooth muscle cells (SMC) were treated with anti‐TGF‐β antibody, angiotensin II (AngII), anti‐TGF‐β antibody + AngII, or isotype control. ROS were detected during early aneurysm formation in the Fbn1C1039G/+ AS aorta, but absent in normal‐sized DES aorta. Fbn1C1039G/+ mice treated with the unspecific NADPH oxidase inhibitor, apocynin reduced AS aneurysm formation, with attenuated elastin fragmentation. In situ zymography revealed apocynin treatment decreased protease activity. In vitro SMC studies showed Fbn1C1039G/+‐derived AS SMC had increased NADPH activity compared to DES‐derived SMC. AS SMC NADPH activity increased with AngII treatment and appeared TGF‐β dependent. In conclusion, ROS play a role in MFS aneurysm development and correspond anatomically with aneurysmal aortic segments. ROS inhibition via apocynin treatment attenuates MFS aneurysm progression. AngII enhances ROS production in MFS AS SMCs and is likely TGF‐β dependent.
Collapse
Affiliation(s)
- Fabian Emrich
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California.,Department of Cardiothoracic Surgery, Leipzig University Heart Center, Leipzig, Germany
| | - Kiril Penov
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California.,Department of Cardiothoracic Surgery, Leipzig University Heart Center, Leipzig, Germany
| | - Mamoru Arakawa
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California.,Department of Cardiovascular Surgery, Jichi Medical University, Saitama, Japan
| | - Nathan Dhablania
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Grayson Burdon
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Albert J Pedroza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Tiffany K Koyano
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Young M Kim
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Uwe Raaz
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | | | - Cristiana Iosef
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| |
Collapse
|
22
|
Pharmacotherapies for the Treatment of Eosinophilic Esophagitis: State of the Art Review. Drugs 2019; 79:1419-1434. [DOI: 10.1007/s40265-019-01173-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Sangha GS, Busch A, Acuna A, Berman AG, Phillips EH, Trenner M, Eckstein HH, Maegdefessel L, Goergen CJ. Effects of Iliac Stenosis on Abdominal Aortic Aneurysm Formation in Mice and Humans. J Vasc Res 2019; 56:217-229. [PMID: 31272099 DOI: 10.1159/000501312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/04/2019] [Indexed: 12/23/2022] Open
Abstract
Reduced lower-limb blood flow has been shown to lead to asymmetrical abdominal aortic aneurysms (AAAs) but the mechanism of action is not fully understood. Therefore, small animal ultrasound (Vevo2100, FUJIFILM VisualSonics) was used to longitudinally study mice that underwent standard porcine pancreatic elastase (PPE) infusion (n = 5), and PPE infusion with modified 20% iliac artery stenosis in the left (n = 4) and right (n = 5) iliac arteries. Human AAA computed tomography images were obtained from patients with normal (n = 9) or stenosed left (n = 2), right (n = 1), and bilateral (n = 1) iliac arteries. We observed rapid early growth and rightward expansion (8/9 mice) in the modified PPE groups (p < 0.05), leading to slightly larger and asymmetric AAAs compared to the standard PPE group. Further examination showed a significant increase in TGFβ1 (p < 0.05) and cellular infiltration (p < 0.05) in the modified PPE group versus standard PPE mice. Congruent, yet variable, observations were made in human AAA patients with reduced iliac outflow compared to those with normal iliac outflow. Our results suggest that arterial stenosis at the time of aneurysm induction leads to faster AAA growth with aneurysm asymmetry and increased vascular inflammation after 8 weeks, indicating that moderate iliac stenosis may have upstream effects on AAA progression.
Collapse
Affiliation(s)
- Gurneet S Sangha
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, Indiana, USA
| | - Albert Busch
- Technical University Munich, Department for Vascular and Endovascular Surgery, Munich Aortic Center, Klinikum rechts der Isar, Munich, Germany
| | - Andrea Acuna
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, Indiana, USA
| | - Alycia G Berman
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, Indiana, USA
| | - Evan H Phillips
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, Indiana, USA
| | - Matthias Trenner
- Technical University Munich, Department for Vascular and Endovascular Surgery, Munich Aortic Center, Klinikum rechts der Isar, Munich, Germany
| | - Hans-Henning Eckstein
- Technical University Munich, Department for Vascular and Endovascular Surgery, Munich Aortic Center, Klinikum rechts der Isar, Munich, Germany
| | - Lars Maegdefessel
- Technical University Munich, Department for Vascular and Endovascular Surgery, Munich Aortic Center, Klinikum rechts der Isar, Munich, Germany
| | - Craig J Goergen
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, Indiana, USA, .,Purdue University, Purdue University Center for Cancer Research, West Lafayette, Indiana, USA,
| |
Collapse
|
24
|
Abstract
The term epidermolysis bullosa (EB) refers to a group of hereditary skin blistering diseases. The group is clinically and genetically heterogeneous, but all EB forms are associated with mechanically induced skin blistering and fragility. The causative gene mutations of most EB types are known. The current international consensus classification contains four main types: EB simplex (EBS), junctional EB (JEB), dystrophic EB (DEB), and Kindler syndrome (KS). The classification is based on the morphological level of blister formation. In EBS, the split is intra-epidermal, in JEB along the basement membrane and in DEB below the basement membrane. In Kindler syndrome, the dermal-epidermal junction is disorganized, and blisters can occur on all three levels. Each major EB type has further subtypes which may differ in terms of their genetic, biological or clinical characteristics. Traditionally, EB treatments have been symptomatic, but increasing understanding of disease etio-pathogenesis is facilitating development of novel evidence-based therapy approaches. First gene- and cell-based therapies are being tested at preclinical level and in clinical trials. New knowledge on secondary disease mechanisms has led to development and clinical testing of urgently needed symptom-relief therapies using small molecules and biologicals.
Collapse
Affiliation(s)
- Leena Bruckner-Tuderman
- Department of Dermatology, Medical Center - University of Freiburg, Hauptstrasse 7, Freiburg 79104, Baden-Wuerttemberg, Germany
| |
Collapse
|
25
|
Wu L. The pathogenesis of thoracic aortic aneurysm from hereditary perspective. Gene 2018; 677:77-82. [DOI: 10.1016/j.gene.2018.07.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 01/19/2023]
|
26
|
Yao Y, Li Y, Zeng X, Ye Z, Li X, Zhang L. Losartan Alleviates Renal Fibrosis and Inhibits Endothelial-to-Mesenchymal Transition (EMT) Under High-Fat Diet-Induced Hyperglycemia. Front Pharmacol 2018; 9:1213. [PMID: 30420805 PMCID: PMC6215973 DOI: 10.3389/fphar.2018.01213] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022] Open
Abstract
The endothelial-to-mesenchymal transition (EMT) of glomerular vascular endothelial cells is considered to be pivotal in diabetic nephropathy (DN). The risk of DN can be decreased by losartan, but the potential molecular mechanism(s) are not fully understood. Extensive data show that the EMT occurs in proximal tubular endothelial cells resulting in an endothelial phenotype switch (fibrotic matrix accumulation), consequently enhancing the development of renal interstitial fibrosis. Here, we found that losartan significantly ameliorated DN-induced renal fibrosis progression via inhibition of the EMT in mice. In vivo experiments suggested that losartan significantly alleviated microalbuminuria and pathologic changes under high-fat diet-induced hyperglycemia. Immunohistochemistry indicated that losartan suppressed the EMT in glomeruli. In addition, losartan decreased oxidative stress damage and inhibited the transforming growth factor (TGF)-β1/Smad pathway. Furthermore, consistent changes were detected in vitro where losartan markedly inhibited the EMT and TGF-β1/Smad pathway induced by high glucose in glomerular endothelial cells. Together, these results suggested that losartan could alleviate the EMT in glomeruli via inhibition of oxidative stress damage and the TGF-β1/Smad signaling pathway under hyperglycemia.
Collapse
Affiliation(s)
- Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Zeng
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Zheng Ye
- College of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xia Li
- Institute of Agricultural Resources and Environment, Guangdong Academy of Agricultural Sciences, Guangdong, China
| | - Lu Zhang
- Wenhua College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Kimura N, Futamura K, Arakawa M, Okada N, Emrich F, Okamura H, Sato T, Shudo Y, Koyano TK, Yamaguchi A, Adachi H, Matsuda A, Kawahito K, Matsumoto K, Fischbein MP. Gene expression profiling of acute type A aortic dissection combined with in vitro assessment. Eur J Cardiothorac Surg 2018; 52:810-817. [PMID: 28402522 DOI: 10.1093/ejcts/ezx095] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/26/2017] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The mechanisms underlying aortic dissection remain to be fully elucidated. We aimed to identify key molecules driving dissection through gene expression profiling achieved by microarray analysis and subsequent in vitro experiments using human aortic endothelial cells (HAECs) and aortic vascular smooth muscle cells (AoSMCs). METHODS Total RNA, including microRNA (miRNA), was isolated from the intima-media layer of dissected ascending aorta obtained intraoperatively from acute type A aortic dissection (ATAAD) patients without familial thoracic aortic disease (n = 8) and that of non-dissected ascending aorta obtained from transplant donors (n = 9). Gene expression profiling was performed with mRNA and miRNA microarrays, and results were confirmed by quantitative polymerase chain reaction (qPCR). Target genes and miRNA were identified by gene ontology analysis and a literature search. To reproduce the in silico results, HAECs and AoSMCs were stimulated in vitro by upstream cytokines, and expression of target genes was assessed by qPCR. RESULTS Microarray analysis revealed 1536 genes (3.6%, 1536/42 545 probes) and 41 miRNAs (3.0%, 41/1368 probes) that were differentially expressed in the ATAAD group (versus donor group). The top 15 related pathways included regulation of inflammatory response, growth factor activity and extracellular matrix. Gene ontology analysis identified JAK2 (regulation of inflammatory response), PDGFA, TGFB1, VEGFA (growth factor activity) and TIMP3, TIMP4, SERPINE1 (extracellular matrix) as the target genes and miR-21-5p, a TIMP3 repressor, as target miRNA that interacts with the target genes. Validation qPCR confirmed the altered expression of all 7 target genes and miR-21-5p in dissected aorta specimens (all genes, P < 0.05). Ingenuity pathway analysis showed TNF-α and TGF-β to be upstream cytokines for the target genes. In vitro experiments showed these cytokines inhibit TIMP3 expression (P < 0.05) and enhance VEGFA expression (P < 0.01) in AoSMCs but not HAECs. miR-21-5p expression increases in AoSMCs under TNF-α and TGF-β stimulation (fold change: 1.36; P = 0.011). CONCLUSIONS Results of our novel approach, integrating in vitro assessment into gene expression profiling, implicated chronic inflammation characterized by MMP-TIMP dysregulation, increased VEGFA expression, and TGF-β signalling in the development of dissection. Further investigation may reveal novel diagnostic biomarkers and uncover the mechanism(s) underlying ATAAD.
Collapse
Affiliation(s)
- Naoyuki Kimura
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Kyoko Futamura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Mamoru Arakawa
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Naoko Okada
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Fabian Emrich
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Homare Okamura
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Tetsuya Sato
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Tiffany K Koyano
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| | - Atsushi Yamaguchi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Hideo Adachi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Akio Matsuda
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Koji Kawahito
- Division of Cardiovascular Surgery, Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University, School of Medicine, Stanford, CA, USA
| |
Collapse
|
28
|
Lin CJ, Lin CY, Stitziel NO. Genetics of the extracellular matrix in aortic aneurysmal diseases. Matrix Biol 2018; 71-72:128-143. [PMID: 29656146 DOI: 10.1016/j.matbio.2018.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/17/2022]
Abstract
Aortic aneurysms are morbid conditions that can lead to rupture or dissection and are categorized as thoracic (TAA) or abdominal aortic aneurysms (AAA) depending on their location. While AAA shares overlapping risk factors with atherosclerotic cardiovascular disease, TAA exhibits strong heritability. Human genetic studies in the past two decades have successfully identified numerous genes involved in both familial and sporadic forms of aortic aneurysm. In this review we will discuss the genetic basis of aortic aneurysm, focusing on the extracellular matrix and how insights from these studies have informed our understanding of human biology and disease pathogenesis.
Collapse
Affiliation(s)
- Chien-Jung Lin
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Chieh-Yu Lin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; McDonell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
29
|
Kusters PJH, Seijkens TTP, Beckers L, Lievens D, Winkels H, de Waard V, Duijvestijn A, Lindquist Liljeqvist M, Roy J, Daugherty A, Newby A, Gerdes N, Lutgens E. CD40L Deficiency Protects Against Aneurysm Formation. Arterioscler Thromb Vasc Biol 2018. [PMID: 29519940 DOI: 10.1161/atvbaha.117.310640] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The mechanisms underlying formation of arterial aneurysms remain incompletely understood. Because inflammation is a common feature during the progressive degeneration of the aortic wall, we studied the role of the costimulatory molecule CD40L, a major driver of inflammation, in aneurysm formation. APPROACH AND RESULTS Transcriptomics data obtained from human abdominal aortic aneurysms and normal aortas revealed increased abundance of both CD40L and CD40 in media of thrombus-free and thrombus-covered human abdominal aortic aneurysms samples. To further unravel the role of CD40L in aneurysm formation, apolipoprotein E-deficient (Apoe-/-) and Cd40l-/-Apoe-/- mice were infused with angiotensin II for 7 and 28 days. Only a minority of Cd40l-/-Apoe-/- mice (33% and 17%) developed (dissecting) aneurysms compared with 75% and 67% of Apoe-/- littermates after 7 and 28 days of infusion, respectively. Total vessel area of the aorta at the suprarenal level was 52% smaller in angiotensin II-infused Cd40l-/-Apoe-/- mice compared with that in angiotensin II-infused Apoe-/- mice. Chimeric Apoe-/- mice repopulated with Cd40l-/-Apoe-/- bone marrow afforded a similar protection against dissecting aneurysm formation. Moreover, lack of CD40L protected mice from fatal aneurysm rupture. T helper cell and macrophage accumulation in aneurysmal tissue was reduced in Cd40l-/-Apoe-/- mice with a concomitant decrease in expression of proinflammatory chemo- and cytokines. In addition, aneurysms of Cd40l-/-Apoe-/- mice displayed reduced abundance of matrix metalloproteinase-13 and an increase in tissue inhibitor of metalloproteinase-3 while activity of matrix metalloproteinase-2 and matrix metalloproteinase-9 was diminished. CONCLUSIONS Deficiency of (hematopoietic) CD40L protects against dissecting aneurysm formation and reduces the incidence of fatal rupture. This is associated with a decreased accumulation and activation of inflammatory cells and a dampened protease activity in the arterial wall.
Collapse
Affiliation(s)
- Pascal J H Kusters
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.)
| | - Tom T P Seijkens
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.).,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany (T.T.P.S., D.L., H.W., N.G., E.L.)
| | - Linda Beckers
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.)
| | - Dirk Lievens
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.).,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany (T.T.P.S., D.L., H.W., N.G., E.L.)
| | - Holger Winkels
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.).,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany (T.T.P.S., D.L., H.W., N.G., E.L.)
| | - Vivian de Waard
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.)
| | | | - Moritz Lindquist Liljeqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (M.L.L., J.R.)
| | - Joy Roy
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (M.L.L., J.R.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Andrew Newby
- Bristol Heart Institute, University of Bristol, United Kingdom (A.N.)
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany (T.T.P.S., D.L., H.W., N.G., E.L.).,Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Germany (N.G.)
| | - Esther Lutgens
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (P.J.H.K., T.T.P.S., L.B., D.L., H.W., V.d.W., E.L.) .,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany (T.T.P.S., D.L., H.W., N.G., E.L.)
| |
Collapse
|
30
|
Chiarelli N, Carini G, Zoppi N, Ritelli M, Colombi M. Transcriptome analysis of skin fibroblasts with dominant negative COL3A1 mutations provides molecular insights into the etiopathology of vascular Ehlers-Danlos syndrome. PLoS One 2018; 13:e0191220. [PMID: 29346445 PMCID: PMC5773204 DOI: 10.1371/journal.pone.0191220] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/29/2017] [Indexed: 01/20/2023] Open
Abstract
Vascular Ehlers-Danlos syndrome (vEDS) is a dominantly inherited connective tissue disorder caused by mutations in the COL3A1 gene that encodes type III collagen (COLLIII), which is the major expressed collagen in blood vessels and hollow organs. The majority of disease-causing variants in COL3A1 are glycine substitutions and in-frame splice mutations in the triple helix domain that through a dominant negative effect are associated with the severe clinical spectrum potentially lethal of vEDS, characterized by fragility of soft connective tissues with arterial and organ ruptures. To shed lights into molecular mechanisms underlying vEDS, we performed gene expression profiling in cultured skin fibroblasts from three patients with different structural COL3A1 mutations. Transcriptome analysis revealed significant changes in the expression levels of several genes involved in maintenance of cell redox and endoplasmic reticulum (ER) homeostasis, COLLs folding and extracellular matrix (ECM) organization, formation of the proteasome complex, and cell cycle regulation. Protein analyses showed that aberrant COLLIII expression is associated with the disassembly of many structural ECM constituents, such as fibrillins, EMILINs, and elastin, as well as with the reduction of the proteoglycans perlecan, decorin, and versican, all playing an important role in the vascular system. Furthermore, the altered distribution of the ER marker protein disulfide isomerase PDI and the strong reduction of the COLLs-modifying enzyme FKBP22 are consistent with the disturbance of ER-related homeostasis and COLLs biosynthesis and post-translational modifications, indicated by microarray analysis. Our findings add new insights into the pathophysiology of this severe vascular disorder, since they provide a picture of the gene expression changes in vEDS skin fibroblasts and highlight that dominant negative mutations in COL3A1 also affect post-translational modifications and deposition into the ECM of several structural proteins crucial to the integrity of soft connective tissues.
Collapse
Affiliation(s)
- Nicola Chiarelli
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Giulia Carini
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Nicoletta Zoppi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Marco Ritelli
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Marina Colombi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
- * E-mail:
| |
Collapse
|
31
|
Falvella FS, Marelli S, Cheli S, Montanelli S, Viecca F, Salvi L, Ferrara A, Clementi E, Trifirò G, Pini A. Pharmacogenetic approach to losartan in Marfan patients: a starting point to improve dosing regimen? Drug Metab Pers Ther 2017; 31:157-63. [PMID: 27474842 DOI: 10.1515/dmpt-2016-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/01/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND Losartan is under evaluation for managing Marfan patients with aortic root dilatation. Cytochrome P450 (CYP) enzymes convert losartan to E3174 active metabolite. The aim of this study is to describe the distribution of CYP2C9*2, CYP2C9*3, CYP3A4*22 and CYP3A5*3 defective alleles, according to losartan tolerance in paediatric Marfan patients. METHODS We genotyped 53 paediatric Marfan patients treated with losartan. The rate of aortic root dilatation was evaluated using the delta z-score variation. Differences in tolerated losartan daily doses with respect to CYP metabolic classes were assessed through the Kruskal-Wallis test. RESULTS The losartan daily dose spans from 0.16 to 2.50 mg/kg (median 1.10 mg/kg). As we expect from the pharmacokinetics pathway, we observe highest tolerated dose in CYP2C9 poor metabolisers (median 1.50 mg/kg, interquartile range 1.08-1.67 mg/kg); however, this difference is not statistically significant. CONCLUSIONS The optimal dose of angiotensin receptor blocker is not known, and no data are available about losartan pharmacogenetic profile in Marfan syndrome; we have proposed a strategy to tackle this issue based on evaluating the major genetic polymorphisms involved in the losartan conversion into active carboxylic acid metabolite. Further studies are needed to support the use of genetic polymorphisms as predictors of the right dose of losartan.
Collapse
|
32
|
MacFarlane EG, Haupt J, Dietz HC, Shore EM. TGF-β Family Signaling in Connective Tissue and Skeletal Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022269. [PMID: 28246187 DOI: 10.1101/cshperspect.a022269] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transforming growth factor β (TGF-β) family of signaling molecules, which includes TGF-βs, activins, inhibins, and numerous bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), has important functions in all cells and tissues, including soft connective tissues and the skeleton. Specific TGF-β family members play different roles in these tissues, and their activities are often balanced with those of other TGF-β family members and by interactions with other signaling pathways. Perturbations in TGF-β family pathways are associated with numerous human diseases with prominent involvement of the skeletal and cardiovascular systems. This review focuses on the role of this family of signaling molecules in the pathologies of connective tissues that manifest in rare genetic syndromes (e.g., syndromic presentations of thoracic aortic aneurysm), as well as in more common disorders (e.g., osteoarthritis and osteoporosis). Many of these diseases are caused by or result in pathological alterations of the complex relationship between the TGF-β family of signaling mediators and the extracellular matrix in connective tissues.
Collapse
Affiliation(s)
- Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia Haupt
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,Howard Hughes Medical Institute, Bethesda, Maryland 21205
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
33
|
Yamashiro Y, Yanagisawa H. Crossing Bridges between Extra- and Intra-Cellular Events in Thoracic Aortic Aneurysms. J Atheroscler Thromb 2017; 25:99-110. [PMID: 28943527 PMCID: PMC5827090 DOI: 10.5551/jat.rv17015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thoracic aortic aneurysms (TAAs) are common, life-threatening diseases and are a major cause of mortality and morbidity. Over the past decade, genetic approaches have revealed that 1) activation of the transforming growth factor beta (TGF-β) signaling, 2) alterations in the contractile apparatus of vascular smooth muscle cells (SMCs), and 3) defects in the extracellular matrix (ECM) were responsible for development of TAAs. Most recently, a fourth mechanism has been proposed in that dysfunction of mechanosensing in the aortic wall in response to hemodynamic stress may be a key driver of TAAs. Interestingly, the elastin-contractile unit, which is an anatomical and functional unit connecting extracellular elastic laminae to the intracellular SMC contractile filaments, via cell surface receptors, has been shown to play a critical role in the mechanosensing of SMCs, and many genes identified in TAAs encode for proteins along this continuum. However, it is still debated whether these four pathways converge into a common pathway. Currently, an effective therapeutic strategy based on the underlying mechanism of each type of TAAs has not been established. In this review, we will update the present knowledge on the molecular mechanism of TAAs with a focus on the signaling pathways potentially involved in the initiation of TAAs. Finally, we will evaluate current therapeutic strategies for TAAs and propose new directions for future treatment of TAAs.
Collapse
Affiliation(s)
- Yoshito Yamashiro
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba
| | - Hiromi Yanagisawa
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba
| |
Collapse
|
34
|
Abstract
Marfan syndrome is a connective tissue disorder that can affect many organ systems. Affected patients present with orthopaedic manifestations of the syndrome during all phases of life. Pain caused by musculoskeletal abnormalities often requires definitive orthopaedic treatment. Orthopaedic surgeons must understand the phenotypes of Marfan syndrome so they can recognize when screening is warranted and can appropriately address the skeletal manifestations. Through medical advancements, patients with Marfan syndrome are living longer and more active lives. Knowledge of the latest diagnostic criteria for the disorder, as well as of advances in understanding the skeletal phenotype, clinical trials of medication therapy, and lifestyle considerations is important for orthopaedic surgeons who treat these patients because these clinicians often are the first to suspect Marfan syndrome and recommend screening.
Collapse
|
35
|
Liu R, Lo L, Lay AJ, Zhao Y, Ting KK, Robertson EN, Sherrah AG, Jarrah S, Li H, Zhou Z, Hambly BD, Richmond DR, Jeremy RW, Bannon PG, Vadas MA, Gamble JR. ARHGAP18 Protects Against Thoracic Aortic Aneurysm Formation by Mitigating the Synthetic and Proinflammatory Smooth Muscle Cell Phenotype. Circ Res 2017; 121:512-524. [DOI: 10.1161/circresaha.117.310692] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Rationale:
Thoracic aortic aneurysm (TAA) is a potentially lethal condition, which can affect individuals of all ages. TAA may be complicated by the sudden onset of life-threatening dissection or rupture. The underlying mechanisms leading to TAA formation, particularly in the nonsyndromal idiopathic group of patients, are not well understood. Thus, identification of new genes and targets that are involved in TAA pathogenesis are required to help prevent and reverse the disease phenotype.
Objective:
Here we explore the role of ARHGAP18, a novel Rho GAP expressed by smooth muscle cells (SMCs), in the pathogenesis of TAA.
Methods and Results:
Using human and mouse aortic samples, we report that ARHGAP18 levels were significantly reduced in the SMC layer of aortic aneurysms.
Arhgap18
global knockout (
Arhgap18
−/
−
) mice exhibited a highly synthetic, proteolytic, and proinflammatory smooth muscle phenotype under basal conditions and when challenged with angiotensin II, developed TAA with increased frequency and severity compared with littermate controls. Chromatin immunoprecipitation studies revealed this phenotype is partly associated with strong enrichment of H3K4me3 and depletion of H3K27me3 at the
MMP2
and
TNF-α
promoters in
Arhgap18
-deficient SMC. We further show that TAA formation in the
Arhgap18
−/−
mice is associated with loss of Akt activation. The abnormal SMC phenotype observed in the
Arhgap18
−/−
mice can be partially rescued by pharmacological treatment with the mTORC1 inhibitor rapamycin, which reduces the synthetic and proinflammatory phenotype of
Arhgap18
-deficient SMC.
Conclusion:
We have identified
ARHGAP18
as a novel protective gene against TAA formation and define an additional target for the future development of treatments to limit TAA pathogenesis.
Collapse
Affiliation(s)
- Renjing Liu
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Lisa Lo
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Angelina J. Lay
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Yang Zhao
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Ka Ka Ting
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Elizabeth N. Robertson
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Andrew G. Sherrah
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Sorour Jarrah
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Haibo Li
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Zhaoxiong Zhou
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Brett D. Hambly
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - David R. Richmond
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Richmond W. Jeremy
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Paul G. Bannon
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Mathew A. Vadas
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| | - Jennifer R. Gamble
- From the Agnes Ginges Laboratory for Diseases of the Aorta, Vascular Biology Program (R.L., L.L., H.L.) and Centre for the Endothelium, Vascular Biology Program (A.J.L., Y.Z., K.K.T., S.J., Z.Z., M.A.V., J.R.G.), Centenary Institute, Camperdown, New South Wales, Australia; Discipline of Pathology and Bosch Institute, Charles Perkins Center (E.N.R., B.D.H.) and Sydney Medical School (R.L., E.N.R., R.W.J., P.G.B., M.A.V., J.R.G.), University of Sydney, New South Wales, Australia; The Baird Institute
| |
Collapse
|
36
|
Soiberman U, Foster JW, Jun AS, Chakravarti S. Pathophysiology of Keratoconus: What Do We Know Today. Open Ophthalmol J 2017; 11:252-261. [PMID: 28932341 PMCID: PMC5585454 DOI: 10.2174/1874364101711010252] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/01/2017] [Accepted: 06/14/2017] [Indexed: 12/19/2022] Open
Abstract
Keratoconus is a common corneal ectasia that leads to progressive visual impairment. Numerous studies have shown abnormal protein expression patterns in keratoconic corneas. However, the specific mechanisms causing this disease remain ambiguous. This review aims to provide an update on morphological studies of the keratoconic cornea, relate these early studies with current findings from proteomic, biochemical and cell culture studies and to postulate possible pathogenic pathways.
Collapse
Affiliation(s)
- Uri Soiberman
- Cornea Division, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA
| | - James W Foster
- Cornea Division, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Albert S Jun
- Cornea Division, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Shukti Chakravarti
- Cornea Division, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
37
|
Muppala S, Xiao R, Krukovets I, Verbovetsky D, Yendamuri R, Habib N, Raman P, Plow E, Stenina-Adognravi O. Thrombospondin-4 mediates TGF-β-induced angiogenesis. Oncogene 2017; 36:5189-5198. [PMID: 28481870 PMCID: PMC5589494 DOI: 10.1038/onc.2017.140] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 03/24/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
TGF-β is a multifunctional cytokine affecting many cell types and implicated in tissue remodeling processes. Due to its many functions and cell-specific effects, the consequences of TGF-β signaling are process-and stage-dependent, and it is not uncommon that TGF-β exerts distinct and sometimes opposing effects on a disease progression depending on the stage and on the pathological changes associated with the stage. The mechanisms underlying cell- and process-specific effects of TGF-β are poorly understood. We are describing a novel pathway that mediates induction of angiogenesis in response to TGF-β1. We found that in endothelial cells (EC) TSP-4, a secreted extracellular matrix (ECM) protein is upregulated in response to TGF-β1 and mediates the effects of TGF-β1 on angiogenesis. Upregulation of TSP-4 does not require the synthesis of new protein, is not caused by decreased secretion of TSP-4, and is mediated by activation of SMAD3. Using Thbs4−/− mice and TSP-4 shRNA, we found that TSP-4 mediated pro-angiogenic functions on cultured EC and angiogenesis in vivo in response to TGF-β1. We observed ~ 3-fold increases in tumor mass and levels of angiogenesis markers in animals injected with TGF-β1, and these effects did not occur in Thbs4−/− animals. Injections of an inhibitor of TGF-β1 signaling SB431542 also decreased the weights of tumors and cancer angiogenesis. Our results from in vivo angiogenesis models and cultured EC document that TSP-4 mediates upregulation of angiogenesis by TGF-β1. Upregulation of pro-angiogenic TSP-4 and selective effects of TSP-4 on EC may contribute to stimulation of tumor growth by TGF-β despite the inhibition of cancer cell proliferation.
Collapse
Affiliation(s)
- S Muppala
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - R Xiao
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - I Krukovets
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - D Verbovetsky
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - R Yendamuri
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - N Habib
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | - P Raman
- Department of Integrative Medical Sciences, North Ohio Medical University, Rootstown, OH, USA
| | - E Plow
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
38
|
Okamura H, Emrich F, Trojan J, Chiu P, Dalal AR, Arakawa M, Sato T, Penov K, Koyano T, Pedroza A, Connolly AJ, Rabinovitch M, Alvira C, Fischbein MP. Long-term miR-29b suppression reduces aneurysm formation in a Marfan mouse model. Physiol Rep 2017; 5:5/8/e13257. [PMID: 28455451 PMCID: PMC5408287 DOI: 10.14814/phy2.13257] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022] Open
Abstract
Aortic root aneurysm formation and subsequent dissection and/or rupture remain the leading cause of death in patients with Marfan syndrome. Our laboratory has reported that miR‐29b participates in aortic root/ascending aorta extracellular matrix remodeling during early aneurysm formation in Fbn1C1039G/+ Marfan mice. Herein, we sought to determine whether miR‐29b suppression can reduce aneurysm formation long‐term. Fbn1C1039G/+ Marfan mice were treated with retro‐orbital LNA‐anti‐miR‐29b inhibitor or scrambled‐control‐miR before aneurysms develop either (1) a single dose prenatally (pregnant Fbn1C1039G/+ mice at 14.5 days post‐coitum) (n = 8–10, each group) or (2) postnatally every other week, from 2 to 22 weeks of age, and sacrificed at 24 weeks (n = 8–10, each group). To determine if miR‐29b blockade was beneficial even after aneurysms develop, a third group of animals were treated every other week, starting at 8 weeks of age, until sacrificed (n = 4–6, each group). miR‐29b inhibition resulted in aneurysm reduction, increased elastogenesis, decreased matrix metalloproteinase activity and decreased elastin breakdown. Prenatal LNA‐anti‐miR‐29b inhibitor treatment decreased aneurysm formation up to age 32 weeks, whereas postnatal treatment was effective up to 16 weeks. miR‐29b blockade did not slow aortic growth once aneurysms already developed. Systemic miR‐29b inhibition significantly reduces aneurysm development long‐term in a Marfan mouse model. Drug administration during aortic wall embryologic development appears fundamental. miR‐29b suppression could be a potential therapeutic target for reducing aneurysm formation in Marfan syndrome patients.
Collapse
Affiliation(s)
- Homare Okamura
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Fabian Emrich
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Jeffrey Trojan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Peter Chiu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Alex R Dalal
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Mamoru Arakawa
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Tetsuya Sato
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Kiril Penov
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Tiffany Koyano
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Albert Pedroza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | | | | | - Cristina Alvira
- Department of Pediatrics, Stanford University, Stanford, California
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| |
Collapse
|
39
|
Nitric oxide mediates aortic disease in mice deficient in the metalloprotease Adamts1 and in a mouse model of Marfan syndrome. Nat Med 2017; 23:200-212. [PMID: 28067899 DOI: 10.1038/nm.4266] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022]
Abstract
Heritable thoracic aortic aneurysms and dissections (TAAD), including Marfan syndrome (MFS), currently lack a cure, and causative mutations have been identified for only a fraction of affected families. Here we identify the metalloproteinase ADAMTS1 and inducible nitric oxide synthase (NOS2) as therapeutic targets in individuals with TAAD. We show that Adamts1 is a major mediator of vascular homeostasis, given that genetic haploinsufficiency of Adamts1 in mice causes TAAD similar to MFS. Aortic nitric oxide and Nos2 levels were higher in Adamts1-deficient mice and in a mouse model of MFS (hereafter referred to as MFS mice), and Nos2 inactivation protected both types of mice from aortic pathology. Pharmacological inhibition of Nos2 rapidly reversed aortic dilation and medial degeneration in young Adamts1-deficient mice and in young or old MFS mice. Patients with MFS showed elevated NOS2 and decreased ADAMTS1 protein levels in the aorta. These findings uncover a possible causative role for the ADAMTS1-NOS2 axis in human TAAD and warrant evaluation of NOS2 inhibitors for therapy.
Collapse
|
40
|
Abstract
Thoracic aortic aneurysm is a potentially life-threatening condition in that it places patients at risk for aortic dissection or rupture. However, our modern understanding of the pathogenesis of thoracic aortic aneurysm is quite limited. A genetic predisposition to thoracic aortic aneurysm has been established, and gene discovery in affected families has identified several major categories of gene alterations. The first involves mutations in genes encoding various components of the transforming growth factor beta (TGF-β) signaling cascade (FBN1, TGFBR1, TGFBR2, TGFB2, TGFB3, SMAD2, SMAD3 and SKI), and these conditions are known collectively as the TGF-β vasculopathies. The second set of genes encode components of the smooth muscle contractile apparatus (ACTA2, MYH11, MYLK, and PRKG1), a group called the smooth muscle contraction vasculopathies. Mechanistic hypotheses based on these discoveries have shaped rational therapies, some of which are under clinical evaluation. This review discusses published data on genes involved in thoracic aortic aneurysm and attempts to explain divergent hypotheses of aneurysm origin.
Collapse
Affiliation(s)
- Eric M Isselbacher
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Christian Lacks Lino Cardenas
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark E Lindsay
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
41
|
Wilson NK, Gould RA, Gallo MacFarlane E, Consortium ML. Pathophysiology of aortic aneurysm: insights from human genetics and mouse models. Pharmacogenomics 2016; 17:2071-2080. [PMID: 27922338 DOI: 10.2217/pgs-2016-0127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aneurysms are local dilations of an artery that predispose the vessel to sudden rupture. They are often asymptomatic and undiagnosed, resulting in a high mortality rate. The predisposition to develop thoracic aortic aneurysms is often genetically inherited and associated with syndromes affecting connective tissue homeostasis. This review discusses how elucidation of the genetic causes of syndromic forms of thoracic aortic aneurysm has helped identify pathways that contribute to disease progression, including those activated by TGF-β, angiotensin II and Notch ligands. We also discuss how pharmacological manipulation of these signaling pathways has provided further insight into the mechanism of disease and identified compounds with therapeutic potential in these and related disorders.
Collapse
Affiliation(s)
- Nicole K Wilson
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Russell A Gould
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | | |
Collapse
|
42
|
Ishida J, Konishi M, Ebner N, Springer J. Repurposing of approved cardiovascular drugs. J Transl Med 2016; 14:269. [PMID: 27646033 PMCID: PMC5029061 DOI: 10.1186/s12967-016-1031-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/07/2016] [Indexed: 12/19/2022] Open
Abstract
Research and development of new drugs requires both long time and high costs, whereas safety and tolerability profiles make the success rate of approval very low. Drug repurposing, applying known drugs and compounds to new indications, has been noted recently as a cost-effective and time-unconsuming way in developing new drugs, because they have already been proven safe in humans. In this review, we discuss drug repurposing of approved cardiovascular drugs, such as aspirin, beta-blockers, angiotensin converting enzyme inhibitors, angiotensin II receptor blockers, cardiac glycosides and statins. Regarding anti-tumor activities of these agents, a number of experimental studies have demonstrated promising pleiotropic properties, whereas all clinical trials have not shown expected results. In pathological conditions other than cancer, repurposing of cardiovascular drugs is also expanding. Numerous experimental studies have reported possibilities of drug repurposing in this field and some of them have been tried for new indications ('bench to bedside'), while unexpected results of clinical studies have given hints for drug repurposing and some unknown mechanisms of action have been demonstrated by experimental studies ('bedside to bench'). The future perspective of experimental and clinical studies using cardiovascular drugs are also discussed.
Collapse
Affiliation(s)
- Junichi Ishida
- Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Centre Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Masaaki Konishi
- Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Centre Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Nicole Ebner
- Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Centre Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Jochen Springer
- Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Centre Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| |
Collapse
|
43
|
Frikeche J, Maiti G, Chakravarti S. Small leucine-rich repeat proteoglycans in corneal inflammation and wound healing. Exp Eye Res 2016; 151:142-9. [PMID: 27569372 DOI: 10.1016/j.exer.2016.08.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
The small leucine rich repeat proteoglycans are major components of the cornea. Lumican, keratocan, decorin, biglycan and osteoglycin are present throughout the adult corneal stroma, and fibromodulin in the peripheral limbal area. In the cornea literature these proteoglycan have been reviewed as structural, collagen fibril-regulating proteins of the cornea. However, these proteoglycans are members of the leucine-rich-repeat superfamily, and share structural similarities with pathogen recognition toll-like receptors. Emerging studies are showing that these have a range of interactions with cell surface receptors, chemokines, growth factors and pathogen associated molecular patterns and are able to regulate host immune response, inflammation and wound healing. This review discusses what is known about their innate immune-related role directly in the cornea, and studies outside the field that find interesting links with innate immune and wound healing responses that are likely to be relevant to the ocular surface. In addition, the review discusses phenotypes of mice with targeted deletion of proteoglycan genes and genetic variants associated with human pathologies.
Collapse
Affiliation(s)
- Jihane Frikeche
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - George Maiti
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Shukti Chakravarti
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, USA; Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, USA.
| |
Collapse
|
44
|
Robertson EN, Hambly BD, Jeremy RW. Thoracic aortic dissection and heritability: forensic implications. Forensic Sci Med Pathol 2016; 12:366-8. [PMID: 27393688 DOI: 10.1007/s12024-016-9788-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Elizabeth N Robertson
- Sydney Medical School, University of Sydney, Sydney, NSW, 2008, Australia.
- Discipline of Pathology, Level 4W, Charles Perkins Centre (D17), University of Sydney, Sydney, NSW, 2008, Australia.
| | - Brett D Hambly
- Discipline of Pathology, Level 4W, Charles Perkins Centre (D17), University of Sydney, Sydney, NSW, 2008, Australia
| | - Richmond W Jeremy
- Sydney Medical School, University of Sydney, Sydney, NSW, 2008, Australia
| |
Collapse
|
45
|
Nyström A, Thriene K, Mittapalli V, Kern JS, Kiritsi D, Dengjel J, Bruckner-Tuderman L. Losartan ameliorates dystrophic epidermolysis bullosa and uncovers new disease mechanisms. EMBO Mol Med 2016. [PMID: 26194911 PMCID: PMC4568953 DOI: 10.15252/emmm.201505061] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Genetic loss of collagen VII causes recessive dystrophic epidermolysis bullosa (RDEB)—a severe skin fragility disorder associated with lifelong blistering and disabling progressive soft tissue fibrosis. Causative therapies for this complex disorder face major hurdles, and clinical implementation remains elusive. Here, we report an alternative evidence-based approach to ameliorate fibrosis and relieve symptoms in RDEB. Based on the findings that TGF-β activity is elevated in injured RDEB skin, we targeted TGF-β activity with losartan in a preclinical setting. Long-term treatment of RDEB mice efficiently reduced TGF-β signaling in chronically injured forepaws and halted fibrosis and subsequent fusion of the digits. In addition, proteomics analysis of losartan- vs. vehicle-treated RDEB skin uncovered changes in multiple proteins related to tissue inflammation. In line with this, losartan reduced inflammation and diminished TNF-α and IL-6 expression in injured forepaws. Collectively, the data argue that RDEB fibrosis is a consequence of a cascade encompassing tissue damage, TGF-β-mediated inflammation, and matrix remodeling. Inhibition of TGF-β activity limits these unwanted outcomes and thereby substantially ameliorates long-term symptoms.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Kerstin Thriene
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany ZBSA Center for Biological Systems Analysis, Freiburg, Germany FRIAS Freiburg Institute for Advanced Studies, Freiburg, Germany
| | - Venugopal Mittapalli
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Johannes S Kern
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Dimitra Kiritsi
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jörn Dengjel
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany ZBSA Center for Biological Systems Analysis, Freiburg, Germany FRIAS Freiburg Institute for Advanced Studies, Freiburg, Germany BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany FRIAS Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
46
|
Flanagan-Steet H, Aarnio M, Kwan B, Guihard P, Petrey A, Haskins M, Blanchard F, Steet R. Cathepsin-Mediated Alterations in TGFß-Related Signaling Underlie Disrupted Cartilage and Bone Maturation Associated With Impaired Lysosomal Targeting. J Bone Miner Res 2016; 31:535-48. [PMID: 26404503 PMCID: PMC4808492 DOI: 10.1002/jbmr.2722] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 11/11/2022]
Abstract
Hypersecretion of acid hydrolases is a hallmark feature of mucolipidosis II (MLII), a lysosomal storage disease caused by loss of carbohydrate-dependent lysosomal targeting. Inappropriate extracellular action of these hydrolases is proposed to contribute to skeletal pathogenesis, but the mechanisms that connect hydrolase activity to the onset of disease phenotypes remain poorly understood. Here we link extracellular cathepsin K activity to abnormal bone and cartilage development in MLII animals by demonstrating that it disrupts the balance of TGFß-related signaling during chondrogenesis. TGFß-like Smad2,3 signals are elevated and BMP-like Smad1,5,8 signals reduced in both feline and zebrafish MLII chondrocytes and osteoblasts, maintaining these cells in an immature state. Reducing either cathepsin K activity or expression of the transcriptional regulator Sox9a in MLII zebrafish significantly improved phenotypes. We further identify components of the large latent TGFß complex as novel targets of cathepsin K at neutral pH, providing a possible mechanism for enhanced Smad2,3 activation in vivo. These findings highlight the complexity of the skeletal disease associated with MLII and bring new insight to the role of secreted cathepsin proteases in cartilage development and growth factor regulation.
Collapse
Affiliation(s)
| | - Megan Aarnio
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Brian Kwan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | | | - Aaron Petrey
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mark Haskins
- Departments of Pathology and Clinical Studies, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | | | - Richard Steet
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| |
Collapse
|
47
|
A dual strategy to improve the penetration and treatment of breast cancer by combining shrinking nanoparticles with collagen depletion by losartan. Acta Biomater 2016; 31:186-196. [PMID: 26675124 DOI: 10.1016/j.actbio.2015.12.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/12/2015] [Accepted: 12/01/2015] [Indexed: 01/24/2023]
Abstract
Although development of nanomedicines has been a promising direction in tumor treatment, the therapeutic outcome of current nanomedicines is unsatisfying, partly because of the poor retention and penetration in tumors. Recently, a kind of tumor microenvironment sensitive size shrinkable nanoparticles (DOX-AuNPs-GNPs) has been developed by our lab, which could enhance the tumor penetration and retention depending on the size shrinking. However, the further enhancement is still restricted by dense collagen network in tumors. Thus in this study, we combined DOX-AuNPs-GNPs with losartan to deplete tumor collagen (constituted up to 90% of extracellular matrix) to further improve tumor penetration. In vitro, DOX-AuNPs-GNPs can shrink from over 117.8nm to less than 50.0nm and release DOX-AuNPs under the triggering of tumor overexpressed matrix metalloproteinases-2 (MMP-2). In vivo, pretreatment with losartan significantly decrease the collagen level and improve the tumor penetration. In combination, losartan combined with DOX-AuNPs-GNPs showed the best drug delivery efficiency, striking penetration efficiency and best 4T1 breast tumor inhibition effect. In conclusion, this study provided a promising synergetic strategy to improve the tumor treatment efficiency of nanomedicines. STATEMENT OF SIGNIFICANCE We have developed a dual strategy for deep tumor penetration through combining size shrinkable DOX-AuNPs-GNPs with depleting tumor collagen by losartan. Additionally, we demonstrate therapeutic efficacy in breast tumor bearing mouse model. DOX-AuNPs-GNPs co-administration with losartan is a novel and highly attractive strategy for anti-tumor drug delivery with the potential for broad applications in clinic.
Collapse
|
48
|
Singh MN, Lacro RV. Recent Clinical Drug Trials Evidence in Marfan Syndrome and Clinical Implications. Can J Cardiol 2015; 32:66-77. [PMID: 26724512 DOI: 10.1016/j.cjca.2015.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 01/08/2023] Open
Abstract
Marfan syndrome is a genetic disorder of connective tissue with principal manifestations in the cardiovascular, ocular, and skeletal systems. Cardiovascular disease, mainly progressive aortic root dilation and aortic dissection, is the leading cause of morbidity and mortality. The primary aims of this report were to examine the evidence related to medical therapy for Marfan syndrome, including recently completed randomized clinical trials on the efficacy of β-blockers and angiotensin II receptor blockers for the prophylactic treatment of aortic enlargement in Marfan syndrome, and to provide recommendations for medical therapy on the basis of available evidence. Medical therapy for Marfan syndrome should be individualized according to patient tolerance and risk factors such as age, aortic size, and family history of aortic dissection. The Pediatric Heart Network trial showed that atenolol and losartan each reduced the rate of aortic dilation. All patients with known or suspected Marfan syndrome and aortic root dilation should receive medical therapy with adequate doses of either β-blocker or angiotensin receptor blocker. The Pediatric Heart Network trial also showed that atenolol and losartan are more effective at reduction of aortic root z score in younger subjects, which suggests that medical therapy should be prescribed even in the youngest children with aortic dilation. For patients with Marfan syndrome without aortic dilation, the available evidence is less clear. If aortic dilation is severe and/or progressive, therapy with a combination of β-blocker and angiotensin receptor blocker should be considered, although trial results are mixed with respect to the efficacy of combination therapy vs monotherapy.
Collapse
Affiliation(s)
- Michael N Singh
- Department of Cardiology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronald V Lacro
- Department of Cardiology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
49
|
Miyashita T, Miki K, Kamigaki T, Makino I, Nakagawara H, Tajima H, Takamura H, Kitagawa H, Fushida S, Ahmed AK, Duncan MD, Harmon JW, Ohta T. Low-dose gemcitabine induces major histocompatibility complex class I-related chain A/B expression and enhances an antitumor innate immune response in pancreatic cancer. Clin Exp Med 2015; 17:19-31. [PMID: 26449615 DOI: 10.1007/s10238-015-0394-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/12/2015] [Indexed: 02/06/2023]
Abstract
We investigated the effect of gemcitabine (GEM), a key drug for pancreatic cancer treatment, on the expression of cell surface MICA/B in pancreatic cancer cells and resulting cytotoxicity of γδ T cells. We assessed the effect of GEM on the upregulation of cell surface MICA/B expression by flow cytometry, utilizing six pancreatic cancer cell lines. MICA and CD16 expressions from resected pancreatic cancer patient specimens, which received neoadjuvant chemotherapy (NAC) with GEM, were analyzed by immunohistochemistry. GEM could increase MICA/B expression on cell surface in pancreatic cancer cell lines (in 2 of 6 cell lines). This effect was most effectively at concentration not affecting cell growth of GEM (0.001 μM), because MICA/B negative population was appeared at concentration at cytostatic and cytotoxic effect to cell growth (0.1 and 10 μM). The cytotoxic activity of γδ T cells against PANC-1 was detected and functions through interactions between NKG2D and MICA/B. However, the enhancement of NKG2D-dependent cytotoxicity with increased MICA/B expression, by GEM treatment, was not observed. In addition, soluble MIC molecules were released from pancreatic cancer cell lines in culture supernatant with GEM treatment. Immunohistochemical staining demonstrated that MICA expression in tumor cells and CD16 positive cells surrounding tumors were significantly higher in the NAC group compared to that of the control group. There was a significant correlation between NAC and MICA expression, as well as NAC and CD16 positive cell expression. The present results indicate that low-dose GEM-induced MICA/B expression enhances innate immune function rather than cytotoxicity in pancreatic cancer. In addition, our result suggests that the inhibition of cleavage and release of MIC molecules from the tumor surface could potentially improve NKG2D-dependent cytotoxicity.
Collapse
Affiliation(s)
- Tomoharu Miyashita
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Kenji Miki
- Medinet Medical Institute, MEDINET Co., Ltd., 2-2-8 Tamagawadai, Setagaya-ku, Tokyo, 158-0096, Japan
| | - Takashi Kamigaki
- Medinet Medical Institute, MEDINET Co., Ltd., 2-2-8 Tamagawadai, Setagaya-ku, Tokyo, 158-0096, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hisatoshi Nakagawara
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hirohisa Kitagawa
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Ali K Ahmed
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - Mark D Duncan
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - John W Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
50
|
Intrinsic cardiomyopathy in Marfan syndrome: results from in-vivo and ex-vivo studies of the Fbn1C1039G/+ model and longitudinal findings in humans. Pediatr Res 2015; 78:256-63. [PMID: 26042521 DOI: 10.1038/pr.2015.110] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/03/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mild intrinsic cardiomyopathy in patients with Marfan syndrome (MFS) has consistently been evidenced by independent research groups. So far, little is known about the long-term evolution and pathophysiology of this finding. METHODS To gain more insights into the pathophysiology of MFS-related cardiomyopathy, we performed in-vivo and ex-vivo studies of 11 Fbn1(C1039G/+) mice and 9 wild-type (WT) littermates. Serial ultrasound findings obtained in mice were correlated to the human phenotype. We therefore reassessed left ventricular (LV) function parameters over a 6-y follow-up period in 19 previously reported MFS patients, in whom we documented mild LV dysfunction. RESULTS Fbn1(C1039G/+) mice demonstrated LV contractile dysfunction. Subsequent ex-vivo studies of the myocardium of adult mutant mice revealed upregulation of TGFβ-related pathways and consistent abnormalities of the microfibrillar network, implicating a role for microfibrils in the mechanical properties of the myocardium. Echocardiographic parameters did not indicate clinical significant deterioration of LV function during follow-up in our patient cohort. CONCLUSION In analogy with what is observed in the majority of MFS patients, the Fbn1(C1039G/+) mouse model demonstrates mild intrinsic LV dysfunction. Both extracellular matrix and molecular alterations are implicated in MFS-related cardiomyopathy. This model may now enable us to study therapeutic interventions on the myocardium in MFS.
Collapse
|