101
|
Thioredoxin 1 is upregulated in the bone and bone marrow following experimental myocardial infarction: evidence for a remote organ response. Histochem Cell Biol 2020; 155:89-99. [PMID: 33161477 PMCID: PMC7847876 DOI: 10.1007/s00418-020-01939-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 10/31/2022]
Abstract
Ischemia and reperfusion events, such as myocardial infarction (MI), are reported to induce remote organ damage severely compromising patient outcomes. Tissue survival and functional restoration relies on the activation of endogenous redox regulatory systems such as the oxidoreductases of the thioredoxin (Trx) family. Trxs and peroxiredoxins (Prxs) are essential for the redox regulation of protein thiol groups and for the reduction of hydrogen peroxide, respectively. Here, we determined whether experimental MI induces changes in Trxs and Prxs in the heart as well as in secondary organs. Levels and localization of Trx1, TrxR1, Trx2, Prx1, and Prx2 were analyzed in the femur, vertebrae, and kidneys of rats following MI or sham surgery. Trx1 levels were significantly increased in the heart (P = 0.0017) and femur (P < 0.0001) of MI animals. In the femur and lumbar vertebrae, Trx1 upregulation was detected in bone-lining cells, osteoblasts, megakaryocytes, and other hematopoietic cells. Serum levels of Trx1 increased significantly 2 days after MI compared to sham animals (P = 0.0085). Differential regulation of Trx1 in the bone was also detected by immunohistochemistry 1 month after MI. N-Acetyl-cysteine treatment over a period of 1 month induced a significant reduction of Trx1 levels in the bone of MI rats compared to sham and to MI vehicle. This study provides first evidence that MI induces remote organ upregulation of the redox protein Trx1 in the bone, as a response to ischemia-reperfusion injury in the heart.
Collapse
|
102
|
Sayers JR, Riley PR. Heart regeneration: beyond new muscle and vessels. Cardiovasc Res 2020; 117:727-742. [PMID: 33241843 DOI: 10.1093/cvr/cvaa320] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
The most striking consequence of a heart attack is the loss of billions of heart muscle cells, alongside damage to the associated vasculature. The lost cardiovascular tissue is replaced by scar formation, which is non-functional and results in pathological remodelling of the heart and ultimately heart failure. It is, therefore, unsurprising that the heart regeneration field has centred efforts to generate new muscle and blood vessels through targeting cardiomyocyte proliferation and angiogenesis following injury. However, combined insights from embryological studies and regenerative models, alongside the adoption of -omics technology, highlight the extensive heterogeneity of cell types within the forming or re-forming heart and the significant crosstalk arising from non-muscle and non-vessel cells. In this review, we focus on the roles of fibroblasts, immune, conduction system, and nervous system cell populations during heart development and we consider the latest evidence supporting a function for these diverse lineages in contributing to regeneration following heart injury. We suggest that the emerging picture of neurologically, immunologically, and electrically coupled cell function calls for a wider-ranging combinatorial approach to heart regeneration.
Collapse
Affiliation(s)
- Judy R Sayers
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
103
|
de Dios E, Rios-Navarro C, Perez-Sole N, Gavara J, Marcos-Garces V, Rodríguez E, Carratalá A, Forner MJ, Navarro J, Blasco ML, Bondia E, Signes-Costa J, Vila JM, Forteza MJ, Chorro FJ, Bodi V. Similar Clinical Course and Significance of Circulating Innate and Adaptive Immune Cell Counts in STEMI and COVID-19. J Clin Med 2020; 9:jcm9113484. [PMID: 33126723 PMCID: PMC7692467 DOI: 10.3390/jcm9113484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
This study aimed to assess the time course of circulating neutrophil and lymphocyte counts and their ratio (NLR) in ST-segment elevation myocardial infarction (STEMI) and coronavirus disease (COVID)-19 and explore their associations with clinical events and structural damage. Circulating neutrophil, lymphocyte and NLR were sequentially measured in 659 patients admitted for STEMI and in 103 COVID-19 patients. The dynamics detected in STEMI (within a few hours) were replicated in COVID-19 (within a few days). In both entities patients with events and with severe structural damage displayed higher neutrophil and lower lymphocyte counts. In both scenarios, higher maximum neutrophil and lower minimum lymphocyte counts were associated with more events and more severe organ damage. NLR was higher in STEMI and COVID-19 patients with the worst clinical and structural outcomes. A canonical deregulation of the immune response occurs in STEMI and COVID-19 patients. Boosted circulating innate (neutrophilia) and depressed circulating adaptive immunity (lymphopenia) is associated with more events and severe organ damage. A greater understanding of these critical illnesses is pivotal to explore novel alternative therapies.
Collapse
Affiliation(s)
- Elena de Dios
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Cesar Rios-Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | - Nerea Perez-Sole
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | - Jose Gavara
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | | | - Enrique Rodríguez
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Biochemical Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Arturo Carratalá
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Biochemical Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Maria J. Forner
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Internal Medicine Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Jorge Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Medical Directory, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Maria L. Blasco
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Medical Intensive Care Unit, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Elvira Bondia
- Pneumology Service, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Jaime Signes-Costa
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Pneumology Service, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Jose M. Vila
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Physiology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Maria J. Forteza
- Cardiovascular Medicine Unit, Center of Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden;
| | - Francisco J. Chorro
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Vicente Bodi
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain;
- Correspondence: ; Tel.: +34-96-197-3523
| |
Collapse
|
104
|
Ministrini S, Carbone F, Montecucco F. Emerging role for the inflammatory biomarker osteopontin in adverse cardiac remodeling. Biomark Med 2020; 14:1303-1306. [PMID: 33054343 DOI: 10.2217/bmm-2020-0455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/31/2020] [Indexed: 01/07/2023] Open
Affiliation(s)
- Stefano Ministrini
- Internal Medicine, Department of Medicine, Università degli Studi di Perugia, 1 piazzale Gambuli, 06129 Perugia, Italy
- First Clinic of internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| |
Collapse
|
105
|
Calcagno DM, Ng RP, Toomu A, Zhang C, Huang K, Aguirre AD, Weissleder R, Daniels LB, Fu Z, King KR. The myeloid type I interferon response to myocardial infarction begins in bone marrow and is regulated by Nrf2-activated macrophages. Sci Immunol 2020; 5:5/51/eaaz1974. [PMID: 32978242 DOI: 10.1126/sciimmunol.aaz1974] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
Sterile tissue injury is thought to locally activate innate immune responses via damage-associated molecular patterns (DAMPs). Whether innate immune pathways are remotely activated remains relatively unexplored. Here, by analyzing ~145,000 single-cell transcriptomes at steady state and after myocardial infarction (MI) in mice and humans, we show that the type I interferon (IFN) response, characterized by expression of IFN-stimulated genes (ISGs), begins far from the site of injury, in neutrophil and monocyte progenitors within the bone marrow. In the peripheral blood of patients, we observed defined subsets of ISG-expressing neutrophils and monocytes. In the bone marrow and blood of mice, ISG expression was detected in neutrophils and monocytes and their progenitors, intensified with maturation at steady-state and after MI, and was controlled by Tet2 and Irf3 transcriptional regulators. Within the infarcted heart, ISG-expressing cells were negatively regulated by Nrf2 activation in Ccr2- steady-state cardiac macrophages. Our results show that IFN signaling begins in the bone marrow, implicate multiple transcriptional regulators (Tet2, Irf3, and Nrf2) in governing ISG expression, and provide a clinical biomarker (ISG score) for studying IFN signaling in patients.
Collapse
Affiliation(s)
- David M Calcagno
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Richard P Ng
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Avinash Toomu
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Claire Zhang
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Kenneth Huang
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Aaron D Aguirre
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lori B Daniels
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Zhenxing Fu
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kevin R King
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA. .,Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
106
|
Richart AL, Reddy M, Khalaji M, Natoli AL, Heywood SE, Siebel AL, Lancaster GL, Murphy AJ, Carey AL, Drew BG, Didichenko SA, Navdaev AV, Kingwell BA. Apo AI Nanoparticles Delivered Post Myocardial Infarction Moderate Inflammation. Circ Res 2020; 127:1422-1436. [PMID: 32951519 DOI: 10.1161/circresaha.120.316848] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Decades of research have examined immune-modulatory strategies to protect the heart after an acute myocardial infarction and prevent progression to heart failure but have failed to translate to clinical benefit. OBJECTIVE To determine anti-inflammatory actions of n-apo AI (Apo AI nanoparticles) that contribute to cardiac tissue recovery after myocardial infarction. METHODS AND RESULTS Using a preclinical mouse model of myocardial infarction, we demonstrate that a single intravenous bolus of n-apo AI (CSL111, 80 mg/kg) delivered immediately after reperfusion reduced the systemic and cardiac inflammatory response. N-apo AI treatment lowered the number of circulating leukocytes by 30±7% and their recruitment into the ischemic heart by 25±10% (all P<5.0×10-2). This was associated with a reduction in plasma levels of the clinical biomarker of cardiac injury, cardiac troponin-I, by 52±17% (P=1.01×10-2). N-apo AI reduced the cardiac expression of chemokines that attract neutrophils and monocytes by 60% to 80% and lowered surface expression of integrin CD11b on monocytes by 20±5% (all P<5.0×10-2). Fluorescently labeled n-apo AI entered the infarct and peri-infarct regions and colocalized with cardiomyocytes undergoing apoptosis and with leukocytes. We further demonstrate that n-apo AI binds to neutrophils and monocytes, with preferential binding to the proinflammatory monocyte subtype and partially via SR-BI (scavenger receptor BI). In patients with type 2 diabetes, we also observed that intravenous infusion of the same n-apo AI (CSL111, 80 mg/kg) similarly reduced the level of circulating leukocytes by 12±5% (all P<5.0×10-2). CONCLUSIONS A single intravenous bolus of n-apo AI delivered immediately post-myocardial infarction reduced the systemic and cardiac inflammatory response through direct actions on both the ischemic myocardium and leukocytes. These data highlight the anti-inflammatory effects of n-apo AI and provide preclinical support for investigation of its use for management of acute coronary syndromes in the setting of primary percutaneous coronary interventions.
Collapse
Affiliation(s)
- Adele L Richart
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Medini Reddy
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Mina Khalaji
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Alaina L Natoli
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Sarah E Heywood
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | | | - Graeme L Lancaster
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Andrew L Carey
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.)
| | | | | | - Bronwyn A Kingwell
- Baker Heart and Diabetes Institute, Melbourne, Australia (A.L.R., M.R., M.K., A.L.N., S.E.H., G.L.L., A.J.M., B.G.D., B.A.K.).,Department of Physiology (B.A.K.), Monash University, Melbourne, Australia.,School of Medicine (B.A.K.), Monash University, Melbourne, Australia.,CSL Ltd, Bio21, Parkville, Australia (B.A.K.)
| |
Collapse
|
107
|
Thomas TP, Grisanti LA. The Dynamic Interplay Between Cardiac Inflammation and Fibrosis. Front Physiol 2020; 11:529075. [PMID: 33041853 PMCID: PMC7522448 DOI: 10.3389/fphys.2020.529075] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Heart failure is a leading cause of death worldwide. While there are multiple etiologies contributing to the development of heart failure, all cause result in impairments in cardiac function that is characterized by changes in cardiac remodeling and compliance. Fibrosis is associated with nearly all forms of heart failure and is an important contributor to disease pathogenesis. Inflammation also plays a critical role in the heart and there is a large degree of interconnectedness between the inflammatory and fibrotic response. This review discusses the cellular and molecular mechanisms contributing to inflammation and fibrosis and the interplay between the two.
Collapse
Affiliation(s)
- Toby P Thomas
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| | - Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
108
|
Abstract
Interleukin-15 is a pleotropic factor, capable of modulating metabolism, survival, proliferation, and differentiation in many different cell types. The rationale behind this study relates to previous work demonstrating that IL-15 is a major factor present in stem cell extracts, which protects cardiomyocytes subjected to hypoxic stress in vitro. The objective of this current study was to assess whether administration of IL-15 peptide will also show protective effects in vivo. The data indicate that administration of IL-15 reduces cell death, increases vascularity, decreases scar size, and significantly improves left ventricular ejection fraction in a mouse model of myocardial infarction.
Collapse
|
109
|
Castillo EC, Vázquez-Garza E, Yee-Trejo D, García-Rivas G, Torre-Amione G. What Is the Role of the Inflammation in the Pathogenesis of Heart Failure? Curr Cardiol Rep 2020; 22:139. [PMID: 32910299 PMCID: PMC7481763 DOI: 10.1007/s11886-020-01382-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In heart failure, whether it is associated with reduced or preserved ejection fraction, the immune system is activated and contributes to heart remodeling and impaired function. RECENT FINDINGS Studies indicate that cells of the immune system not only play a role in the pathology but are also critical regulators of heart function. Knowledge about the role of the immune system driving heart failure will lead to the development of new targets to this system, particularly in those patients that, despite the apparent wellness, relapse and worsen. In this review, we will address the diverse mechanisms that trigger inflammation and their impact on heart failure progression.
Collapse
Affiliation(s)
- Elena C. Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - Eduardo Vázquez-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - David Yee-Trejo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garza García, NL Mexico
- Tecnologico de Monterrey, Centro de Medicina Funcional, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garzar García, NL Mexico
| | - Guillermo Torre-Amione
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garza García, NL Mexico
- De Bakey CRC, The Methodist Hospital, Cornell University, Houston, TX USA
| |
Collapse
|
110
|
Vafadarnejad E, Rizzo G, Krampert L, Arampatzi P, Arias-Loza AP, Nazzal Y, Rizakou A, Knochenhauer T, Bandi SR, Nugroho VA, Schulz DJJ, Roesch M, Alayrac P, Vilar J, Silvestre JS, Zernecke A, Saliba AE, Cochain C. Dynamics of Cardiac Neutrophil Diversity in Murine Myocardial Infarction. Circ Res 2020; 127:e232-e249. [PMID: 32811295 DOI: 10.1161/circresaha.120.317200] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE After myocardial infarction, neutrophils rapidly and massively infiltrate the heart, where they promote both tissue healing and damage. OBJECTIVE To characterize the dynamics of circulating and cardiac neutrophil diversity after infarction. METHODS AND RESULTS We employed single-cell transcriptomics combined with cell surface epitope detection by sequencing to investigate temporal neutrophil diversity in the blood and heart after murine myocardial infarction. At day 1, 3, and 5 after infarction, cardiac Ly6G+ (lymphocyte antigen 6G) neutrophils could be delineated into 6 distinct clusters with specific time-dependent patterning and proportions. At day 1, neutrophils were characterized by a gene expression profile proximal to bone marrow neutrophils (Cd177, Lcn2, Fpr1), and putative activity of transcriptional regulators involved in hypoxic response (Hif1a) and emergency granulopoiesis (Cebpb). At 3 and 5 days, 2 major subsets of Siglecfhi (enriched for eg, Icam1 and Tnf) and Siglecflow (Slpi, Ifitm1) neutrophils were found. Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) analysis in blood and heart revealed that while circulating neutrophils undergo a process of aging characterized by loss of surface CD62L and upregulation of Cxcr4, heart infiltrating neutrophils acquired a unique SiglecFhi signature. SiglecFhi neutrophils were absent from the bone marrow and spleen, indicating local acquisition of the SiglecFhi signature. Reducing the influx of blood neutrophils by anti-Ly6G treatment increased proportions of cardiac SiglecFhi neutrophils, suggesting accumulation of locally aged neutrophils. Computational analysis of ligand/receptor interactions revealed putative pathways mediating neutrophil to macrophage communication in the myocardium. Finally, SiglecFhi neutrophils were also found in atherosclerotic vessels, revealing that they arise across distinct contexts of cardiovascular inflammation. CONCLUSIONS Altogether, our data provide a time-resolved census of neutrophil diversity and gene expression dynamics in the mouse blood and ischemic heart at the single-cell level, and reveal a process of local tissue specification of neutrophils in the ischemic heart characterized by the acquisition of a SiglecFhi signature.
Collapse
Affiliation(s)
- Ehsan Vafadarnejad
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Wuerzburg, Germany (E.V., A.-E.S.)
| | - Giuseppe Rizzo
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany.,Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Laura Krampert
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany.,Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | | | - Anahi-Paula Arias-Loza
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany
| | - Yara Nazzal
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Anna Rizakou
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Tim Knochenhauer
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Sourish Reddy Bandi
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Vallery Audy Nugroho
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany.,Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Dirk J J Schulz
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany.,Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Melanie Roesch
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Paul Alayrac
- Université de Paris, PARCC, INSERM, F-75015 Paris, France (P. Alayrac, J.V., J.-S.S.)
| | - Jose Vilar
- Université de Paris, PARCC, INSERM, F-75015 Paris, France (P. Alayrac, J.V., J.-S.S.)
| | | | - Alma Zernecke
- Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Wuerzburg, Germany (E.V., A.-E.S.)
| | - Clément Cochain
- Comprehensive Heart Failure Center Wuerzburg (G.R., L.K., A.-P.A.-L., V.A.N., D.J.J.S., C.C.), University Hospital Wuerzburg, Germany.,Institute of Experimental Biomedicine (G.R., L.K.,Y.N., A.R., T.K., S.R.B., V.A.N., D.J.J.S., M.R., A.Z., C.C.), University Hospital Wuerzburg, Germany
| |
Collapse
|
111
|
Mahtta D, Sudhakar D, Koneru S, Silva GV, Alam M, Virani SS, Jneid H. Targeting Inflammation After Myocardial Infarction. Curr Cardiol Rep 2020; 22:110. [PMID: 32770365 DOI: 10.1007/s11886-020-01358-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Inflammation plays a key role in clearing cellular debris and recovery after acute myocardial infarction (AMI). Dysregulation of or prolonged inflammation may result in adverse cardiac remodeling and major adverse clinical events (MACE). Several pre-clinical studies and moderate sized clinical trials have investigated the role of immunomodulation in improving clinical outcomes in patients with AMI. RECENT FINDINGS Clinical data from the Canakinumab Atherothrombosis Outcome (CANTOS) and Colchicine Cardiovascular Outcomes Trial (COLCOT) have provided encouraging results among patients with AMI. Several other clinical and pre-clinical trials have brought about the prospect of modulating inflammation at various junctures of the inflammatory cascade including inhibition of complement cascade, interleukins, and matrix metalloproteinases. In patients with AMI, modulation of residual inflammation via various inflammatory pathways and mediators may hold promise for further reducing MACE. Learning from current data and understanding the nuances of immunomodulation in AMI are key for future trials and before widespread dissemination of such therapies.
Collapse
Affiliation(s)
- Dhruv Mahtta
- Health Policy, Quality & Informatics Program,, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Deepthi Sudhakar
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Srikanth Koneru
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Guilherme Vianna Silva
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Mahboob Alam
- Division of Cardiovascular Medicine,, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Salim S Virani
- Health Policy, Quality & Informatics Program,, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA
| | - Hani Jneid
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX, USA.
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA.
| |
Collapse
|
112
|
Daseke MJ, Tenkorang-Impraim MAA, Ma Y, Chalise U, Konfrst SR, Garrett MR, DeLeon-Pennell KY, Lindsey ML. Exogenous IL-4 shuts off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to induce neutrophil phagocytosis following myocardial infarction. J Mol Cell Cardiol 2020; 145:112-121. [PMID: 32574573 PMCID: PMC7483959 DOI: 10.1016/j.yjmcc.2020.06.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Macrophages and neutrophils are primary leukocytes involved in the inflammatory response to myocardial infarction (MI). While interleukin (IL)-4 is an in vitro anti-inflammatory stimulus, the MI myocardium does not express a considerable amount of IL-4 but does express IL4 receptors. We hypothesized that continuous exogenous IL-4 infusion starting 24 h after MI would promote a polarization switch in inflammatory cells towards a reparative phenotype. METHODS C57BL/6J male mice (3-6 months of age) were subcutaneously infused with either saline (n = 17) or IL-4 (20 ng/g/day; n = 17) beginning 24 h after MI and evaluated at MI day 3. RESULTS Macrophages and neutrophils were isolated ex vivo from the infarct region and examined. Exogenous IL-4 decreased pro-inflammatory Ccl3, Il12a, Tnfa, and Tgfb1 in neutrophils and increased anti-inflammatory Arg1 and Ym1 in macrophages (all p < .05). Tissue clearance by IL-4 treated neutrophils was not different, while selective phagocytosis of neutrophils doubled in IL-4 treated macrophages (p < .05). Of 24,339 genes examined by RNA-sequencing, 2042 genes were differentially expressed in macrophages from IL-4 stimulated infarct (all FDR p < .05). Pdgfc gene expression was ranked first, increasing 3-fold in macrophages stimulated with IL-4 (p = 1 × 10-9). Importantly, changes in macrophage physiology and transcriptome occurred in the absence of global LV effects. Bone marrow derived monocytes stimulated with mouse recombinant PDGF-CC protein (10 μg/ml) or PDGF-CC blocking antibody (200 ng/ml) did not change Arg1 or Ym1 expression, indicating the in vivo effect of IL-4 to stimulate macrophage anti-inflammatory gene expression was independent of PDGF-CC. CONCLUSIONS Our results indicate that exogenous IL-4 promotes inflammation resolution by turning off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to mediate removal of apoptotic neutrophils.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Mavis A A Tenkorang-Impraim
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA; Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
113
|
Yang D, Liu HQ, Liu FY, Tang N, Guo Z, Ma SQ, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. The Roles of Noncardiomyocytes in Cardiac Remodeling. Int J Biol Sci 2020; 16:2414-2429. [PMID: 32760209 PMCID: PMC7378633 DOI: 10.7150/ijbs.47180] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiac remodeling is a common characteristic of almost all forms of heart disease, including cardiac infarction, valvular diseases, hypertension, arrhythmia, dilated cardiomyopathy and other conditions. It is not merely a simple outcome induced by an increase in the workload of cardiomyocytes (CMs). The remodeling process is accompanied by abnormalities of cardiac structure as well as disturbance of cardiac function, and emerging evidence suggests that a wide range of cells in the heart participate in the initiation and development of cardiac remodeling. Other than CMs, there are numerous noncardiomyocytes (non-CMs) that regulate the process of cardiac remodeling, such as cardiac fibroblasts and immune cells (including macrophages, lymphocytes, neutrophils, and mast cells). In this review, we summarize recent knowledge regarding the definition and significant effects of various non-CMs in the pathogenesis of cardiac remodeling, with a particular emphasis on the involved signaling mechanisms. In addition, we discuss the properties of non-CMs, which serve as targets of many cardiovascular drugs that reduce adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Han-Qing Liu
- Department of Thyroid and Breast, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, RP China
| |
Collapse
|
114
|
Vadivel S, Vincent P, Sekaran S, Visaga Ambi S, Muralidar S, Selvaraj V, Palaniappan B, Thirumalai D. Inflammation in myocardial injury- Stem cells as potential immunomodulators for myocardial regeneration and restoration. Life Sci 2020; 250:117582. [PMID: 32222465 DOI: 10.1016/j.lfs.2020.117582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
Abstract
The ineffective immunosuppressant's and targeted strategies to neutralize inflammatory mediators have worsened the scenario of heart failure and have opened many questions for debate. Stem cell therapy has proven to be a promising approach for treating heart following myocardial infarction (MI). Adult stem cells, induced pluripotent stem cells and embryonic stem cells are possible cell types and have successfully shown to regenerate damaged myocardial tissue in pre-clinical and clinical studies. Current implications of using mesenchymal stem cells (MSCs) owing to their immunomodulatory functions and paracrine effects could serve as an effective alternative treatment option for rejuvenating the heart post MI. The major setback associated with the use of MSCs is reduced cell retention, engraftment and decreased effectiveness. With a few reports on understanding the role of inflammation and its dual effects on the structure and function of heart, this review focuses on these missing insights and further exemplifies the role of MSCs as an alternative therapy in treating the pathological consequences in myocardial infarction (MI).
Collapse
Affiliation(s)
- Sajini Vadivel
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Preethi Vincent
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Vimalraj Selvaraj
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
115
|
Understanding the mechanisms that determine extracellular matrix remodeling in the infarcted myocardium. Biochem Soc Trans 2020; 47:1679-1687. [PMID: 31724697 DOI: 10.1042/bst20190113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023]
Abstract
Myocardial Infarction (MI) initiates a series of wound healing events that begins with up-regulation of an inflammatory response and culminates in scar formation. The extracellular matrix (ECM) is intricately involved in all stages from initial break down of existing ECM to synthesis of new ECM to form the scar. This review will summarize our current knowledge on the processes involved in ECM remodeling after MI and identify the gaps that still need to be filled.
Collapse
|
116
|
Fender AC, Dobrev D. Coronary clot composition after myocardial infarction: Thrombus age matters. IJC HEART & VASCULATURE 2020; 26:100450. [PMID: 32142067 PMCID: PMC7046514 DOI: 10.1016/j.ijcha.2019.100450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 11/01/2022]
Affiliation(s)
- Anke C. Fender
- Institute of Pharmacology, Medical Faculty, University Duisburg-Essen, Germany
| | | |
Collapse
|
117
|
Ischemia/Reperfusion Injury: Pathophysiology, Current Clinical Management, and Potential Preventive Approaches. Mediators Inflamm 2020; 2020:8405370. [PMID: 32410868 PMCID: PMC7204323 DOI: 10.1155/2020/8405370] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/18/2019] [Accepted: 01/03/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial ischemia reperfusion syndrome is a complex entity where many inflammatory mediators play different roles, both to enhance myocardial infarction-derived damage and to heal injury. In such a setting, the establishment of an effective therapy to treat this condition has been elusive, perhaps because the experimental treatments have been conceived to block just one of the many pathogenic pathways of the disease, or because they thwart the tissue-repairing phase of the syndrome. Either way, we think that a discussion about the pathophysiology of the disease and the mechanisms of action of some drugs may shed some clarity on the topic.
Collapse
|
118
|
Liu M, Lutz H, Zhu D, Huang K, Li Z, Dinh PC, Gao J, Zhang Y, Cheng K. Bispecific Antibody Inhalation Therapy for Redirecting Stem Cells from the Lungs to Repair Heart Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 8:2002127. [PMID: 33437573 PMCID: PMC7788635 DOI: 10.1002/advs.202002127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/31/2020] [Indexed: 05/13/2023]
Abstract
Stem cell therapy is a promising strategy for cardiac repair. However, clinical efficacy is hampered by poor cell engraftment and the elusive repair mechanisms of the transplanted stem cells. The lung is a reservoir of hematopoietic stem cells (HSCs) and a major biogenesis site for platelets. A strategy is sought to redirect lung resident stem cells to the injured heart for therapeutic repair after myocardial infarction (MI). To achieve this goal, CD34-CD42b platelet-targeting bispecific antibodies (PT-BsAbs) are designed to simultaneously recognize HSCs (via CD34) and platelets (via CD42b). After inhalation delivery, PT-BsAbs reach the lungs and conjoined HSCs and platelets. Due to the innate injury-finding ability of platelets, PT-BsAbs guide lung HSCs to the injured heart after MI. The redirected HSCs promote endogenous repair, leading to increased cardiac function. The repair mechanism involves angiomyogenesis and inflammation modulation. In addition, the inhalation route is superior to the intravenous route to deliver PT-BsAbs in terms of the HSCs' homing ability and therapeutic benefits. This work demonstrates that this novel inhalable antibody therapy, which harnesses platelets derived from the lungs, contributes to potent stem cell redirection and heart repair. This strategy is safe and effective in a mouse model of MI.
Collapse
Affiliation(s)
- Mengrui Liu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityNorth CarolinaUSA
| | - Halle Lutz
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
| | - Dashuai Zhu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityNorth CarolinaUSA
| | - Ke Huang
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
| | - Zhenhua Li
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityNorth CarolinaUSA
| | - Phuong‐Uyen C. Dinh
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
- Comparative Medicine InstituteNorth Carolina State UniversityNorth CarolinaUSA
| | - Junqing Gao
- Department of CardiologyPutuo HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yi Zhang
- Department of CardiologyShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityNorth CarolinaUSA
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityNorth CarolinaUSA
| |
Collapse
|
119
|
Xu S, Xie F, Tian L, Manno SH, Manno FAM, Cheng SH. Prolonged neutrophil retention in the wound impairs zebrafish heart regeneration after cryoinjury. FISH & SHELLFISH IMMUNOLOGY 2019; 94:447-454. [PMID: 31526847 DOI: 10.1016/j.fsi.2019.09.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/11/2019] [Accepted: 09/14/2019] [Indexed: 06/10/2023]
Abstract
Neutrophils are the first line defenders in the innate immune response, and rapidly migrate to an infected or injured area. Recently, bidirectional migration of neutrophils to the wound and the corresponding functions have become popular research pursuits. In zebrafish larvae, CXCR1/CXCL8 is the predominant chemoattractant pathway to recruit neutrophil to wound, while CXCR2/CXCL8 pathway mediate neutrophil dispersal in wound after injury. Here, we found that both CXCR1/CXCL8 and LTB4/BLT1 signals are activated in zebrafish heart after cryoinjury. And with a CXCR1/2 selective inhibitor (SB225002) treatment, the recruitment of neutrophils was not affected, but reverse migration of neutrophils was inhibited after cryoinjury of heart. We suggested that the neutrophil recruitment to cryoinjured area might be mediated by LTB4/BLT1 signals at the presence of SB225002. Therefore, SB225002 treatment resulted more accumulation and long retention of neutrophils in the injured heart. The long retention of neutrophils in the wound promoted revascularization in the injured heart; however, the AKT/mTOR pathway was inhibited and the regeneration was impaired. Our findings suggest that retention of neutrophils is a well-orchestrated process and might regulate regeneration by the AKT/mTOR pathway.
Collapse
Affiliation(s)
- Shisan Xu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, PR China
| | - Fangjing Xie
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, PR China
| | - Li Tian
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, PR China
| | - Sinai Hc Manno
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, PR China
| | - Francis A M Manno
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, New South Wales, Australia
| | - Shuk Han Cheng
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, PR China; State Key Laboratory of Marine Pollution (SKLMP) at City University of Hong Kong, Hong Kong SAR, PR China; Department of Materials Science and Engineering, College of Science and Engineering, City University of Hong Kong, Hong Kong SAR, PR China.
| |
Collapse
|
120
|
Rhee AJ, Lavine KJ. New Approaches to Target Inflammation in Heart Failure: Harnessing Insights from Studies of Immune Cell Diversity. Annu Rev Physiol 2019; 82:1-20. [PMID: 31658002 DOI: 10.1146/annurev-physiol-021119-034412] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite mounting evidence implicating inflammation in cardiovascular diseases, attempts at clinical translation have shown mixed results. Recent preclinical studies have reenergized this field and provided new insights into how to favorably modulate cardiac macrophage function in the context of acute myocardial injury and chronic disease. In this review, we discuss the origins and roles of cardiac macrophage populations in the steady-state and diseased heart, focusing on the human heart and mouse models of ischemia, hypertensive heart disease, and aortic stenosis. Specific attention is given to delineating the roles of tissue-resident and recruited monocyte-derived macrophage subsets. We also highlight emerging concepts of monocyte plasticity and heterogeneity among monocyte-derived macrophages, describe possible mechanisms by which infiltrating monocytes acquire unique macrophage fates, and discuss the putative impact of these populations on cardiac remodeling. Finally, we discuss strategies to target inflammatory macrophage populations.
Collapse
Affiliation(s)
- Aaron J Rhee
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Kory J Lavine
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; .,Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.,Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
121
|
Extracellular annexin-A1 promotes myeloid/granulocytic differentiation of hematopoietic stem/progenitor cells via the Ca 2+/MAPK signalling transduction pathway. Cell Death Discov 2019; 5:135. [PMID: 31552142 PMCID: PMC6755131 DOI: 10.1038/s41420-019-0215-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 12/14/2022] Open
Abstract
Annexin A1 (AnxA1) modulates neutrophil life span and bone marrow/blood cell trafficking thorough activation of formyl-peptide receptors (FPRs). Here, we investigated the effect of exogenous AnxA1 on haematopoiesis in the mouse. Treatment of C57BL/6 mice with recombinant AnxA1 (rAnxA1) reduced the granulocyte–macrophage progenitor (GMP) population in the bone marrow, enhanced the number of mature granulocytes Gr-1+Mac-1+ in the bone marrow as well as peripheral granulocytic neutrophils and increased expression of mitotic cyclin B1 on hematopoietic stem cells (HSCs)/progenitor cells (Lin−Sca-1+c-Kit+: LSK). These effects were abolished by simultaneous treatment with Boc-2, an FPR pan-antagonist. In in vitro studies, rAnxA1 reduced both HSC (LSKCD90lowFLK-2−) and GMP populations while enhancing mature cells (Gr1+Mac1+). Moreover, rAnxA1 induced LSK cell proliferation (Ki67+), increasing the percentage of cells in the S/G2/M cell cycle phases and reducing Notch-1 expression. Simultaneous treatment with WRW4, a selective FPR2 antagonist, reversed the in vitro effects elicited by rAnxA1. Treatment of LSK cells with rAnxA1 led to phosphorylation of PCLγ2, PKC, RAS, MEK, and ERK1/2 with increased expression of NFAT2. In long-term bone marrow cultures, rAnxA1 did not alter the percentage of LSK cells but enhanced the Gr-1+Mac-1+ population; treatment with a PLC (U73122), but not with a PKC (GF109203), inhibitor reduced rAnxA1-induced phosphorylation of ERK1/2 and Elk1. Therefore, we identify here rAnxA1 as an inducer of HSC/progenitor cell differentiation, favouring differentiation of the myeloid/granulocytic lineage, via Ca2+/MAPK signalling transduction pathways.
Collapse
|