1
|
Dunaway L, Mills W, Eyo U, Isakson B. The Cells of the Vasculature: Advances in the Regulation of Vascular Tone in the Brain and Periphery. Basic Clin Pharmacol Toxicol 2025; 136:e70023. [PMID: 40143606 PMCID: PMC11947641 DOI: 10.1111/bcpt.70023] [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: 11/15/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025]
Abstract
The vasculature is a complex tissue in which multiple cell types coordinate the regulation of tissue perfusion in response to hemodynamic and biochemical signals. Advances in this field are continuing to deepen our understanding of the relative importance of these cell types through the body. In the peripheral vasculature, tone is generated primarily by smooth muscle cells and regulated by endothelial cells, and neurons. In the brain parenchyma, unique cell types including pericytes, perivascular astrocytes and microglia, also contribute to the regulation of arterial and capillary tone. Here, we provide a cell-by-cell review of the regulation of vascular tone and highlight recent advances in the regulation of vascular tone in both the periphery and cerebral vasculature.
Collapse
Affiliation(s)
- Luke S. Dunaway
- Robert M. Berne Cardiovascular Research CenterUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - William A. Mills
- Robert M. Berne Cardiovascular Research CenterUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Brain Immunology & Glia Center, Department of NeuroscienceUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Brain InstituteUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Ukpong B. Eyo
- Robert M. Berne Cardiovascular Research CenterUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Brain Immunology & Glia Center, Department of NeuroscienceUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research CenterUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Department of Molecular Physiology and BiophysicsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| |
Collapse
|
2
|
Fu E, Huang KF, Chang HH, Tseng HH, Yang SSD. Periodontitis Increases Gingival, Serum and Hippocampus β-Amyloid Expressions but Reduces Neurovascular Coupling in Basilar Artery of Rats. J Clin Periodontol 2025; 52:762-772. [PMID: 39780369 DOI: 10.1111/jcpe.14110] [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: 08/13/2024] [Revised: 11/19/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
AIM Neurodegenerative diseases are characterized by early increased beta-amyloid (Aβ) and decreased cerebrovascular reactivity. We investigated Aβ in gingiva, serum or hippocampus and neurovascular reactivity in basilar artery (BA) of periodontitis rats, to test the impact of Aβ on BA vasoreactivity ex vivo. MATERIALS AND METHODS Periodontitis was induced in 32 rats using silk-ligation. Rats were sacrificed at weeks 0, 1, 2 and 4. Gingival TNF-α, IL-1β and Aβ were quantified via immunoblotting. Alveolar bone destruction was examined by micro-computed tomography. Serum and hippocampus Aβ values were measured by enzyme-linked immunosorbent assay and fluorescence staining, respectively. Vasoreactivity was measured by myography on isolated BA. RESULTS From Week 1, gingival TNF-α and IL-1β and bone destruction increased. Gingiva, serum and hippocampus Aβ values increased from Week 2. Nicotine-induced BA relaxation declined from Week 2, while acetylcholine-induced relaxation decreased by Week 4. Bone loss correlated with Aβ and nicotine-induced relaxation. Correlations were observed between Aβs in tissues, between two induced BA relaxations and between Aβ expressions and the induced relaxations. Ex vivo, Aβ reduced nicotine- and isoproterenol-induced relaxations but not electrical stimulated relaxation. CONCLUSIONS Periodontitis may increase Aβ expressions and reduce BA neurovascular reactivity, with Aβ contributing to this abnormal neurovascular coupling.
Collapse
Affiliation(s)
- Earl Fu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Kuo-Feng Huang
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Hsi-Hsien Chang
- Division of Urology, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Medical Foundation, New Taipei, Taiwan, ROC
| | - Hui-Hwa Tseng
- Department of Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Stephen Shei-Dei Yang
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
- Division of Urology, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Medical Foundation, New Taipei, Taiwan, ROC
| |
Collapse
|
3
|
Costa TJ, Fontes MT, Barros PR, Hope MC, Webb RC, Wenceslau CF, Enos RT, McCarthy CG. Overexpression of adipose tissue ERα enhances PVAT anticontractility via NOX4-derived H 2O 2 and is protective against high-fat diet-induced dysfunction. Am J Physiol Heart Circ Physiol 2025; 328:H1065-H1072. [PMID: 40127093 DOI: 10.1152/ajpheart.00180.2025] [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: 03/12/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Menopause has unequivocally been associated with cardiovascular risk and obesity. Loss of estrogen bioavailability is a hallmark of menopause. Estrogen is generally considered vasculoprotective, with estrogen receptor α (ERα) being the predominant receptor subtype that mediates these positive effects. Similarly, estrogen and ERα are known to stimulate white adipose tissue metabolism. However, it is unknown whether ERα could exert a beneficial effect on mesenteric perivascular adipose tissue (PVAT). PVAT is a heterogeneous tissue that surrounds most peripheral blood vessels. In physiological conditions, PVAT has an anticontractile effect on the vasculature. However, in several diseases, PVAT switches its phenotype to become procontractile. To date, the role of ERα in PVAT function in health and disease is unknown. Therefore, we hypothesized that overexpression of adipose tissue ERα (ERαOE) would 1) increase the anticontractile effect of PVAT in chow diet conditions and 2) protect mice against a high-fat diet (HFD)-induced PVAT dysfunction. To test this hypothesis, mesenteric resistance arteries, with and without PVAT, were isolated from female ERαOE mice, which had either been on a regular chow diet or an HFD for 19 wk. We observed that ERαOE amplifies the anticontractile effect of mesenteric PVAT via NADPH oxidase 4 (NOX4)-derived hydrogen peroxide (H2O2) in chow conditions, and ERαOE is protective against a dysfunctional PVAT that is observed after an HFD, via the same anticontractile mechanism. Collectively, these data demonstrate that ERα is vasculoprotective in the context of PVAT. Harnessing this signaling could be important for reducing cardiovascular risk in postmenopausal women.NEW & NOTEWORTHY We have revealed for the first time that overexpression of adipose tissue estrogen receptor α (ERαOE) amplifies the anticontractile effect of mesenteric PVAT via the biosynthesis of NADPH oxidase 4 (NOX4)-derived hydrogen peroxide (H2O2), and this overexpression is protective against HFD-induced PVAT dysfunction. Collectively, these data demonstrate an important mechanism by which ERα signaling is vasculoprotective in the context of PVAT.
Collapse
Affiliation(s)
- Tiago J Costa
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Milene T Fontes
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Paula R Barros
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Marion C Hope
- Department of Pathology, Microbiology, & Immunology, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, United States
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Reilly T Enos
- Department of Pathology, Microbiology, & Immunology, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine-Columbia, Columbia, South Carolina, United States
| |
Collapse
|
4
|
Luse MA, Schug WJ, Dunaway LS, Nyshadham S, Loeb SA, Carvalho A, Tessema R, Pavelic C, Keller TCS, Shu X, Ruddiman CA, Kosmach A, Sveeggen TM, Mitchell R, Bagher P, Minshal RD, Leitnger N, Columbus L, Levental KR, Levental I, Cortese-Krott M, Isakson BE. Nitrosation of CD36 Regulates Endothelial Function and Serum Lipids. Arterioscler Thromb Vasc Biol 2025. [PMID: 40242868 DOI: 10.1161/atvbaha.124.321964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND During obesity, endothelial cells (ECs) become lipid laden, leading to endothelial dysfunction. We tested posttranslational modification on CD36 that may regulate EC lipid accumulation. METHODS We used an EC-specific Cav1 (caveolin-1) knockout mouse, nitrosation and palmitoylation assays, and whole animal Nγ-nitro-l-arginine methyl ester administration to examine blood lipids. RESULTS EC-specific Cav1 knockout male mice are hyperlipidemic regardless of diet but retain endothelial cell function. We found these mice have significantly increased NO in response to the lack of Cav1, and the presence or absence of NO toggled inversely EC lipid content and plasma lipid in mice. The NO nitrosated the fatty acid translocase CD36 at the same cysteines that are palmitoylated on CD36. The nitrosation of CD36 prevented its trafficking to the plasma membrane and decreased lipid accumulation. The physiological effect of this mechanism was a reliance on NO for endothelial function and not dilation. CONCLUSIONS This work suggests that CD36 nitrosation occurs as a protective mechanism to prevent EC lipotoxicity.
Collapse
Affiliation(s)
- Melissa A Luse
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Wyatt J Schug
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Skylar A Loeb
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Alicia Carvalho
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Rachel Tessema
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Caitlin Pavelic
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, China (X.S.)
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Anna Kosmach
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Timothy M Sveeggen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Ray Mitchell
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Pooneh Bagher
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | | | - Norbert Leitnger
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville (L.C.)
| | - Kandice R Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Ilya Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | | | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| |
Collapse
|
5
|
Cho JM, Park SK, Mookherjee S, Peters EC, Pires PW, Symons JD. Bryostatin-1 improves function in arteries with suppressed endothelial cell autophagy. GeroScience 2025:10.1007/s11357-025-01650-5. [PMID: 40220153 DOI: 10.1007/s11357-025-01650-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
We have previously reported that when autophagy is suppressed in endothelial cells (ECs), a glycolytic defect limits shear-stress -induced ATP production to an extent that purinergic 2Y1 receptor (P2Y1R)-mediated activation of EC nitric oxide (NO) synthase (eNOS) is compromised. Subsequently we demonstrated the functional relevance of this finding in arteries from mice with genetic, pharmacological, and age-associated EC autophagy impairment. Using gain and loss of function approaches in vitro, we further revealed that p-PKCδT505 serves as a signaling link between P2Y1R activation and NO generation. Here we sought to discern the functional relevance of this observation. First, shear-stress- induced activating phosphorylation of eNOS (p-eNOSS1177) that is otherwise prevented by knockdown of autophagy-related gene 3 (Atg3) in ECs was restored by the PKC agonist bryostatin-1. Next, in murine models of genetic and age-associated EC autophagy compromise, depressed vasodilation displayed by femoral and cerebral arteries was reversed by bryostatin-1 in a manner that could be prevented by concurrent NO synthase inhibition. Finally, the bryostatin-1-mediated normalization of intraluminal flow-induced vasodilation observed in femoral arteries from both models of EC autophagy disruption was mitigated by inhibiting downstream targets of p-PKCδT505 i.e., p-PKDS744/S748 and p-PKDS916. These findings provide evidence that stimulating PKC/PKD has strategic potential to restore compromised endothelial function in pathologies associated with suppressed EC autophagy e.g., aging.
Collapse
Affiliation(s)
- Jae Min Cho
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA.
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA.
| | - Seul-Ki Park
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Sohom Mookherjee
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona Health Science Center, Tucson, AZ, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
6
|
Prabhahar A, Batta A, Hatwal J, Kumar V, Ramachandran R, Batta A. Endothelial dysfunction in the kidney transplant population: Current evidence and management strategies. World J Transplant 2025; 15:97458. [PMID: 40104196 PMCID: PMC11612885 DOI: 10.5500/wjt.v15.i1.97458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/04/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
The endothelium modulates vascular homeostasis owing to a variety of vasoconstrictors and vasodilators. Endothelial dysfunction (ED), characterized by impaired vasodilation, inflammation, and thrombosis, triggers future cardiovascular (CV) diseases. Chronic kidney disease, a state of chronic inflammation caused by oxidative stress, metabolic abnormalities, infection, and uremic toxins damages the endothelium. ED is also associated with a decline in estimated glomerular filtration rate. After kidney transplantation, endothelial functions undergo immediate but partial restoration, promising graft longevity and enhanced CV health. However, the anticipated CV outcomes do not happen due to various transplant-related and unrelated risk factors for ED, culminating in poor CV health and graft survival. ED in kidney transplant recipients is an under-recognized and poorly studied entity. CV diseases are the leading cause of death among kidney transplant candidates with functioning grafts. ED contributes to the pathogenesis of many of the CV diseases. Various biomarkers and vasoreactivity tests are available to study endothelial functions. With an increasing number of transplants happening every year, and improved graft rejection rates due to the availability of effective immunosuppressants, the focus has now shifted to endothelial protection for the prevention, early recognition, and treatment of CV diseases.
Collapse
Affiliation(s)
- Arun Prabhahar
- Department of Telemedicine (Internal Medicine and Nephrology), Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Akshey Batta
- Department of Urology and Renal Transplant, Neelam Hospital, Rajpura 140401, Punjab, India
| | - Juniali Hatwal
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vivek Kumar
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Raja Ramachandran
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
| |
Collapse
|
7
|
Carvalho KFS, de Lima JF, Silva JLM, de Almeida CR, Cunha RGA, Alves JV, Tostes RC, Lobato NS, Costa RM. Toll-like receptor 9 contributes to perivascular adipose tissue dysfunction in spontaneously hypertensive rats. Eur J Pharmacol 2025; 998:177524. [PMID: 40097130 DOI: 10.1016/j.ejphar.2025.177524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/19/2025]
Abstract
Perivascular adipose tissue (PVAT) plays a key role in vascular homeostasis by exerting anticontractile effects. However, PVAT dysfunction in hypertension contributes to vascular abnormalities via inflammation and oxidative stress. This study investigates the role of Toll-like receptor 9 (TLR9) in PVAT dysfunction in spontaneously hypertensive rats (SHR). Elevated TLR9 expression and activation were observed in SHR PVAT, along with increased pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) and reactive oxygen species (ROS). These changes impaired PVAT's anticontractile effects, reduced nitric oxide (NO) bioavailability, and heightened vascular contraction. Pharmacological inhibition of TLR9 with ODN2088 restored PVAT's anticontractile function, reduced inflammation and oxidative stress, and improved vascular tone. This treatment also significantly lowered systolic blood pressure in SHR. TLR9-mediated PVAT dysfunction was closely linked to NF-κB signaling, as inhibition of this pathway attenuated inflammatory cytokine production and improved vascular reactivity. ROS scavenging with Tiron confirmed the role of oxidative stress in the loss of PVAT function. Despite unaltered endothelial nitric oxide synthase (eNOS) expression, NO levels were reduced in SHR PVAT due to ROS-induced scavenging. Notably, TLR9 inhibition restored NO bioavailability, reinforcing its therapeutic potential. These findings establish TLR9 as a critical mediator of PVAT dysfunction in hypertension, driving inflammation, oxidative stress, and vascular impairment. Targeting TLR9 and oxidative stress may represent effective therapeutic strategies for mitigating vascular dysfunction in hypertension.
Collapse
Affiliation(s)
- Karine F S Carvalho
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - Júlia F de Lima
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - João Lucas M Silva
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | | | - Rosana G A Cunha
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - Juliano V Alves
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Núbia S Lobato
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - Rafael M Costa
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil.
| |
Collapse
|
8
|
Lyall M, Kamdar A, Sykes R, Aekbote BL, Gadegaard N, Berry C. Measuring contractile forces in vascular smooth muscle cells. Vascul Pharmacol 2025; 159:107488. [PMID: 40097082 DOI: 10.1016/j.vph.2025.107488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Vascular smooth muscle cell (VSMC) contractility mediates blood vessel tone. Abnormalities in VSMC function and in blood vessel tone can contribute to a variety of cardiovascular diseases. This review examines the role of VSMC contractile force in vascular disease, divided into two primary sections. The first section introducing VSMC mechanical contraction and detailing the molecular mechanisms of VSMC contractility in normal and pathological states. The second section exploring methods of measuring contraction in VSMCs, such as Ca2+ imaging, myography, and traction force microscopy, and highlighting where each method is of best use. Understanding the mechanical properties and contractile profiles of VSMCs offers valuable insights into disease mechanisms. By investigating these aspects, this review describes the potential of VSMC contractile forces as diagnostic markers and therapeutic targets in vascular disease.
Collapse
Affiliation(s)
- Maia Lyall
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Badri L Aekbote
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK; Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK.
| |
Collapse
|
9
|
He S, Singh D, Helfield B. Flow rate modulates focused ultrasound-mediated vascular delivery of microRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102426. [PMID: 39850318 PMCID: PMC11754011 DOI: 10.1016/j.omtn.2024.102426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/10/2024] [Indexed: 01/25/2025]
Abstract
Gene therapy targeting ischemic heart disease is a promising therapeutic avenue, but it is mostly restricted to viral-based delivery approaches which are limited due to off-target immunological responses. Focused ultrasound presents a non-viral, image-guided technique in which circulating intravascular microbubble contrast agents can reversibly enhance vascular permeability and gene penetration. Here, we explore the influence of flow rate on the microbubble-assisted delivery of miR-126, a potent pro-angiogenic biologic, using a custom acoustically coupled pressurized mesenteric artery model. We demonstrate that under the same ultrasound conditions, increased flow rates enhance microbubble-mediated cell permeability; yet, miR-126 delivery itself exhibits a negative correlation with increasing flow velocity. Post-ultrasound assays confirmed vessel vasoreactivity, maintaining vasoconstriction and vasodilation capacities. These findings underscore the critical role microbubble flow rate plays in focused ultrasound gene therapy, especially notable for applications in which blood velocity itself is a salient pathophysiological indicator of disease progression, including ischemia.
Collapse
Affiliation(s)
- Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Davindra Singh
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada
- Department of Physics, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
10
|
Fellows AL, Quigley K, Leung V, Ainscough AJ, Wilkins MR, Barnett H, Miller D, Mayr M, Wojciak-Stothard B. Engineered pulmonary artery tissues for measuring contractility, drug testing and disease modelling. Br J Pharmacol 2025. [PMID: 39979037 DOI: 10.1111/bph.17462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Vasoreactivity of pulmonary arteries regulates blood flow through the lungs. Excessive constriction of these vessels contributes to pulmonary arterial hypertension (PAH), a progressive and incurable condition, resulting in right heart failure. The search for new and improved drug treatments is hampered by laboratory models that do not reproduce the vasoactive behaviour of healthy and diseased human arteries. EXPERIMENTAL APPROACH We have developed an innovative technique for producing miniature, three-dimensional arterial structures that allow proxy evaluation of human pulmonary artery contractility. These "engineered pulmonary artery tissues" or "EPATs" are fabricated by suspending human pulmonary artery vascular smooth muscle cells (VSMCs) in fibrin hydrogels between pairs of silicone posts, located on custom-made racks, in 24-well culture plates. KEY RESULTS EPATs exhibit rapid, robust and reproducible contraction responses to vasoconstrictors (KCl, ET-1, U46619) as well as relaxation responses to clinically approved PAH vasodilatory drugs that target several signalling pathways, such as bosentan, epoprostenol, selexipag and imatinib. EPATs composed of pulmonary artery VSMCs from PAH patients exhibit enhanced contraction to vasoconstrictors and relaxation in response to vasodilators. We also demonstrate the incorporation of endothelial cells into EPATs for the measurement of endothelium-dependent dilatory responses. CONCLUSION AND IMPLICATIONS We demonstrate the capacity and suitability of EPATs for studying the contractile behaviour of human arterial cells and preclinical drug testing. This novel biomimetic platform has the potential to dramatically improve our understanding and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Adam L Fellows
- National Heart & Lung Institute, Imperial College London, London, UK
- Imperial College Advanced Hackspace, Imperial College London, London, UK
| | - Kate Quigley
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Venus Leung
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Martin R Wilkins
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Harry Barnett
- Imperial College Advanced Hackspace, Imperial College London, London, UK
| | - David Miller
- Imperial College Advanced Hackspace, Imperial College London, London, UK
| | - Manuel Mayr
- National Heart & Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
11
|
Corken A, Weinkopff T, Wahl EC, Sikes JD, Thakali KM. Platelets Modulate Leukocyte Population Composition Within Perivascular Adipose Tissue. Int J Mol Sci 2025; 26:1625. [PMID: 40004089 PMCID: PMC11855773 DOI: 10.3390/ijms26041625] [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: 01/13/2025] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Perivascular adipose tissue (PVAT) regulates vascular tone and is composed of adipocytes and several leukocyte subpopulations. Diet can modify PVAT function, as obesogenic diets cause morphological changes to adipocytes and skew the leukocyte phenotype, leading to PVAT dysregulation and impaired vasoregulation. Of note, platelets, the clot-forming cells, also modulate many facets of leukocyte activity, such as tissue infiltration and polarity. We aimed to determine whether platelets regulate the leukocyte populations residing within PVAT. Male C57Bl/6J mice were fed a Western diet (30% kcal sucrose, 40% kcal fat, 8.0% sodium) to develop obesogenic conditions for PVAT leukocyte remodeling. Diet was either administered acutely (2 weeks) or extended (8 weeks) to gauge the length of challenge necessary for remodeling. Additionally, platelet depletion allowed for the assessment of platelet relevance in PVAT leukocyte remodeling. Abdominal PVAT (aPVAT) and thoracic PVAT (tPVAT) were then isolated and leukocyte composition evaluated by flow cytometry. Compared to control, Western diet alone did not significantly impact PVAT leukocyte composition for either diet length. Platelet depletion, independent of diet, significantly disrupted PVAT leukocyte content with monocytes/macrophages most impacted. Furthermore, tPVAT appeared more sensitive to platelet depletion than aPVAT, providing novel evidence of platelet regulation of leukocyte composition within PVAT depots.
Collapse
Affiliation(s)
- Adam Corken
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Elizabeth C. Wahl
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - James D. Sikes
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - Keshari M. Thakali
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| |
Collapse
|
12
|
Yamakoshi KI, Rolfe P, Yamakoshi T. Arteriolar Elasticity Measurement in the Fingertip Based on Photoplethysmographic Volume-Oscillometry: A New Approach to the Assessment of Vasomotor Functions in the Microvasculature. Cardiovasc Eng Technol 2025:10.1007/s13239-025-00772-3. [PMID: 39825176 DOI: 10.1007/s13239-025-00772-3] [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: 09/02/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025]
Abstract
PURPOSE Dysfunction of vasomotor reactions due to arteriolar smooth muscle causes serious adverse events, such as loss of hemodynamic coherence. This in turn can increase risks of cardiovascular-related diseases. A noninvasive and quantitative evaluation of microvascular disorder is therefore very important for early diagnosis and treatment. This paper describes a new approach to the assessment of vasomotor functions using the arteriolar elasticity measurement technique in the fingertip. METHODS A recently developed device, modified to detect a photoplethysmogram with green light (gPPG) in arteriolar regions, allowed the measurement of arteriolar blood pressure (BPca.) and gPPG from a left index fingertip placed on an occlusive cuff of the device. Arteriolar stiffness and distensibility were analyzed as effective elasticity indices, as a function of arteriolar distending pressure derived by volume-oscillometry. Cold pressor tests to induce vasoconstriction were carried out whether appropriate elasticity changes could be obtained. RESULTS Experiments using 6 healthy subjects were successfully made to obtain arteriolar elastic properties before and while immersing a right hand in cold water. The index-values of stiffness and distensibility showed, respectively, a considerable increase and decrease, clearly demonstrating the appropriate elasticity changes with vasoconstrictive reactions. CONCLUSION Although a further study using many subjects is needed, the results so far suggest that this method could easily provide important features to acquire quantitatively arteriolar elasticity together with BPca. and to assess vasomotor functions in the microvasculature. This convenient method appears useful for clinical practices and health management and promising also for screening cardiovascular-related diseases. (242/250 words).
Collapse
Affiliation(s)
- Ken-Ichi Yamakoshi
- Department of Orthopedic Surgery, Showa University School of Medicine, Shinagawa, Tokyo, 142-8555, Japan
- College of Science and Engineering, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
- Department of Research and Development, Nonprofit Organization of Research Institute of Life Benefit, Sapporo, Hokkaido, 005-0006, Japan
| | - Peter Rolfe
- Department of Automatic Measurement and Control, Harbin Institute of Technology, No. 92 West Dazhi Street, Nan Gang District, Harbin, Heilongjiang Province, 150001, China
- Science and Technology, Oxford BioHorizons, 12 Park View Road, Berkhamsted, UK
| | - Takehiro Yamakoshi
- Department of Research and Development, Nonprofit Organization of Research Institute of Life Benefit, Sapporo, Hokkaido, 005-0006, Japan.
- Institute of Liberal Arts and Science, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
13
|
Saputra F, Tseng TM, Casuga FP, Lai YH, Hung CH, Hsiao CD. Application of a ImageJ-Based Method to Measure Blood Flow in Adult Zebrafish and Its Applications for Toxicological and Pharmacological Assessments. BIOLOGY 2025; 14:51. [PMID: 39857282 PMCID: PMC11763070 DOI: 10.3390/biology14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Blood flow is an important physiological endpoint to measure cardiovascular performance in animals. Because of their innate transparent bodies, zebrafish is an excellent animal model for assessing in vivo cardiovascular performance. Previously, various helpful methods for measuring blood flow in zebrafish larvae were discovered and developed. However, an optimized method to measure blood flow in adult zebrafish has not been reported. In this paper, the tail fin region was selected as target for blood flow measurements using the Trackmate method, provided by ImageJ platform. Based on power statistic calculations, the aortic vessel at the tail base was selected, and other parameters, such as ambient temperature, were investigated for method standardization, in order to minimize experimental variation. The method was also validated using fenpropathrin and ponatinib, which showed some cardiac alterations in a previous zebrafish study. We also checked the versatility of this method by following the same setup in black tetra and medaka and found that this method performed well. However, our results show that heavy pigmentation, like that found in tiger barb, and overlapping vessels, like those in parrot fish, make it hard for this method to perform well. Overall, an optimized protocol was used for the first time to measure blood flow velocity in adult wild-type zebrafish without the aid of transgenic lines or fluorescent dye.
Collapse
Affiliation(s)
- Ferry Saputra
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Orthopaedics, E-Da Hospital/E-Da Dachang Hospital, Kaohsiung 82445, Taiwan
| | - Tzu-Ming Tseng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Franelyne P. Casuga
- Department of Pharmacy, Research Center for the Natural and Applied Science, University of Santo Tomas, Manila 1008, Philippines;
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Chih-Hsin Hung
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| |
Collapse
|
14
|
Gupta K, Llewellyn J, Roberts E, Liu C, Naji A, Assoian RK, Wells RG. Biliary atresia susceptibility gene EFEMP1 regulates extrahepatic bile duct elastic fiber formation and mechanics. JHEP Rep 2025; 7:101215. [PMID: 39717503 PMCID: PMC11663959 DOI: 10.1016/j.jhepr.2024.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 12/25/2024] Open
Abstract
Background & Aims EGF-containing fibulin extracellular matrix protein 1 (EFEMP1, also called fibulin-3) is an extracellular matrix protein linked in a genome-wide association study to biliary atresia, a fibrotic disease of the neonatal extrahepatic bile duct. Fibulin-3 is deposited in most tissues and Efemp1 null mice have decreased elastic fibers in visceral fascia; however, fibulin-3 does not have a role in the development of large elastic fibers and its overall function in the extrahepatic bile ducts remains unclear. Methods We used staining and histology to define the amount and organization of key extracellular matrix components in the extrahepatic bile ducts. We also repurposed pressure myography, a technique heretofore applied to the vasculature, to determine the contribution of elastin and fibulin-3 to extrahepatic bile duct mechanics. We examined extrahepatic bile duct structure and mechanics in three models: neonatal vs. adult rat ducts (n = 6 each), elastase-treated adult rat ducts (n = 6-7 each), and Efemp1 +/- vs. wild-type mouse ducts (n = 6 each). Results We demonstrated that fibulin-3 is expressed in the submucosa of both neonatal and adult mouse, rat and human extrahepatic bile ducts and that, in adult Efemp1 +/- mouse ducts, elastin organization into fibers is decreased by approximately half. Pressure myography showed that Efemp1 +/- ducts have altered mechanics compared to control ducts, with Efemp1 +/- ducts displaying significant stretch compared to controls (p = 0.0376); these changes in stretch are similar to those observed in elastase-treated vs. normal ducts (p <0.0001) and in neonatal ducts vs. adult ducts (p <0.0001). Conclusion Fibulin-3 has an important role in the formation of elastic fibers and the mechanical properties of the extrahepatic bile duct. This provides functional relevance for the biliary atresia susceptibility gene EFEMP1. Impact and implications The gene EFEMP1 was found via a genome-wide association study to be a susceptibility gene for the neonatal disease biliary atresia. EFEMP1 encodes the protein fibulin-3, which regulates elastic fiber organization in the extrahepatic bile duct (EHBD), the major site of disease in biliary atresia. We showed that neonatal EHBDs as well as mice heterozygous for Efemp1 have decreased numbers of elastic fibers, and that this alters EHBD mechanics. This work is important for understanding the mechanism of biliary atresia, in particular susceptibility to obstruction.
Collapse
Affiliation(s)
- Kapish Gupta
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Llewellyn
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Emilia Roberts
- Program in Translational Biomechanics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, PA, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, PA, USA
| | - Richard K. Assoian
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
- Program in Translational Biomechanics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, PA, USA
| | - Rebecca G. Wells
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Jensen LJ. Pressure Myography. Methods Mol Biol 2025; 2894:89-109. [PMID: 39699812 DOI: 10.1007/978-1-0716-4342-6_8] [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] [Indexed: 12/20/2024]
Abstract
Vasomotor function (constriction, dilation) can be assessed ex vivo using the pressure myograph technique, also referred to as perfusion myography in older literature. The technique involves isolating an artery (or any other blood vessel/lymphatic vessel) from an animal research model or from surgery-resected human tissue. The vessel preparation is mounted between two tiny glass pipettes through which a physiological saline solution (usually Krebs') is perfused while superfusing the preparation with the same solution. The inner and outer diameter of the vessel can be measured in real time, giving valuable information about the vessel reactivity in response to various vasoconstrictor or vasodilator stimuli in different research models or treatments. In addition, by measuring the passive (100% relaxed) inner and outer diameters of the vessel, important structural information is obtained, which is used to assess the degree of structural remodeling in a disease model or treatment. This technique is not a high-throughput technique and requires highly dedicated and trained personnel to carry out experiments. The reward, however, is highly physiologically relevant data, which can be used to assess the vascular function/dysfunction in almost any vascularized tissue of any human disease model.
Collapse
Affiliation(s)
- Lars Jørn Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
Wang Z, Mu W, Zhong J, Xu R, Liu Y, Zhao G, Guo Y, Zhang J, Surakka I, Chen YE, Chang L. Vascular smooth muscle cell PRDM16 regulates circadian variation in blood pressure. J Clin Invest 2024; 135:e183409. [PMID: 39625782 PMCID: PMC11785921 DOI: 10.1172/jci183409] [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: 05/30/2024] [Accepted: 11/27/2024] [Indexed: 02/03/2025] Open
Abstract
Disruptions of blood pressure (BP) circadian variation are closely associated with an increased risk of cardiovascular disease. Thus, gaining insights into the molecular mechanisms of BP circadian variation is essential for comprehending BP regulation. Human genetic analyses suggest that PR domain-containing protein 16 (PRDM16), a transcription factor highly expressed in vascular smooth muscle cells (VSMCs), is significantly associated with BP-related traits. However, the roles of PRDM16 in BP regulation are largely unknown. Here, we demonstrate that BP in VSMC-specific Prdm16-KO (Prdm16SMKO) mice was significantly lower than that in control mice during the active period, resulting in aberrant BP circadian variation. Mesenteric artery rings from Prdm16SMKO mice showed a reduced response to phenylephrine. Mechanistically, we identified adrenergic receptor α 1d (Adra1d) as a transcriptional target of PRDM16. Notably, PRDM16 exhibited a remarkable circadian expression pattern and regulated the expression of clock genes, particularly Npas2, which is crucial for BP circadian variation regulation. Consequently, PRDM16 deficiency in VSMCs caused disrupted BP circadian variation through a reduced response to adrenergic signaling and clock gene regulation. Our findings provide insights into the intricate molecular pathways that govern circadian fluctuations in BP.
Collapse
Affiliation(s)
- Zhenguo Wang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjuan Mu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Juan Zhong
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Ruiyan Xu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Department of Pathophysiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yaozhong Liu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Guizhen Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Yanhong Guo
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Ida Surakka
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Smith JA, Ramirez-Perez FI, Burr K, Gonzalez-Vallejo JD, Morales-Quinones M, McMillan NJ, Ferreira-Santos L, Sharma N, Foote CA, Martinez-Lemus LA, Padilla J, Manrique-Acevedo C. Impact of dietary supplementation of glycocalyx precursors on vascular function in type 2 diabetes. J Appl Physiol (1985) 2024; 137:1592-1603. [PMID: 39480270 PMCID: PMC11687847 DOI: 10.1152/japplphysiol.00651.2024] [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: 08/22/2024] [Revised: 10/10/2024] [Accepted: 10/25/2024] [Indexed: 12/10/2024] Open
Abstract
Degradation of the endothelial glycocalyx in type 2 diabetes (T2D) is thought to contribute to impaired shear stress mechanotransduction, leading to endothelial dysfunction and the development of cardiovascular disease. Herein, we tested the hypothesis that restoration of the endothelial glycocalyx with dietary supplementation of glycocalyx precursors (DSGPs, containing glucosamine sulfate, fucoidan, superoxide dismutase, and high-molecular weight hyaluronan) improves endothelial function and other indices of vascular function in T2D. First, in db/db mice, we showed that treatment with DSGP (100 mg/kg/day) for 4 wk restored endothelial glycocalyx length, as assessed via atomic force microscopy in aortic explants. Restoration of the glycocalyx with DSGP was accompanied by improved flow-mediated dilation (FMD) and reduced arterial stiffness in isolated mesenteric arteries. Further corroborating these findings, the treatment of cultured endothelial cells with that same mixture of glycocalyx precursors promoted glycocalyx growth. Next, as an initial step to investigate the translatability of these findings, we conducted a pilot (n = 22) double-blinded randomized placebo-controlled clinical trial to assess the effects of DSGP (3,712.5 mg/day) for 8 wk on endothelial glycocalyx integrity and indices of vascular function, including FMD, in Veterans with T2D. Contrary to the hypothesis, DSGP neither enhanced endothelial glycocalyx integrity nor improved vascular function indices relative to placebo. Together, these findings conceptually support the notion that restoration of the endothelial glycocalyx can lead to improvements in vascular function in a mouse model of T2D; however, DSGP as a therapeutic strategy to enhance vascular function in individuals with T2D does not appear to be efficacious.NEW & NOTEWORTHY Endothelial glycocalyx degradation in type 2 diabetes (T2D) is thought to contribute to impaired shear stress mechanotransduction, leading to vascular dysfunction. The findings of this study support the notion that restoration of the endothelial glycocalyx using a dietary supplementation of glycocalyx precursors can lead to improvements in vascular function in diabetic mice. However, the utilized dietary supplement as a therapeutic strategy to enhance vascular function in individuals with T2D is not efficacious.
Collapse
Affiliation(s)
- James A Smith
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | | | - Katherine Burr
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | | | | | - Neil J McMillan
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | | | - Neekun Sharma
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Christopher A Foote
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
18
|
Flanigan EG, Farman GP, Dennis MR, Wollman L, Van Den Berg M, Granzier H, Banek CT, Fregosi RF. Developmental nicotine exposure alters cardiovascular structure and function in neonatal and juvenile rats. Am J Physiol Heart Circ Physiol 2024; 327:H1442-H1454. [PMID: 39453426 PMCID: PMC11684951 DOI: 10.1152/ajpheart.00558.2024] [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: 08/13/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Here we test the hypothesis that continuous nicotine exposure throughout pre- and postnatal development (developmental nicotine exposure, DNE) alters the cardiovascular structure and function in neonatal and juvenile rats. Echocardiography showed that DNE reduced left ventricular mass, left ventricular outflow tract (LVOT) diameter, and posterior wall thickness, but only in females. Both male and female DNE rats had a lower end-systolic volume, higher ejection fraction, and increased fractional shortening, with unchanged stroke volume and cardiac output. Left ventricular single cardiac myocytes from male and female DNE animals exhibited increased calcium-evoked maximal tension with no effect on EC50. Tail-cuff plethysmography in awake rats showed that DNE males had lower systolic blood pressure and higher heart rate than control males. No significant changes in preload, afterload, or the in vitro renal artery response to vasodilators were observed. The results suggest that DNE enhances myocyte tension-generating capacity, possibly compensating for an unknown developmental insult, which may differ in males and females. Although this adaptation maintains normal resting cardiac function, it may lead to reduced cardiac reserve, increased energy demand, and elevated oxidative stress, potentially compromising both short- and long-term cardiovascular health in developing neonates.NEW & NOTEWORTHY Developmental nicotine exposure (DNE) induced cardiovascular changes in neonatal/juvenile rats. Relative to controls, females had reduced left ventricular mass and dimensions, while both sexes had increased ejection fraction and fractional shortening. DNE increased calcium-evoked tension in cardiac myocytes, suggesting an adaptive mechanism as resting cardiac output was preserved. Despite normal resting function, these changes may reduce cardiac reserve, potentially compromising long-term cardiovascular health. These novel findings highlight how DNE disrupts cardiovascular development and function.
Collapse
Affiliation(s)
- Emily G Flanigan
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Gerrie P Farman
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Melissa R Dennis
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Lila Wollman
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Marloes Van Den Berg
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Henk Granzier
- Department of Cellular & Molecular Medicine, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Christopher T Banek
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| | - Ralph F Fregosi
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
- Department of Neuroscience, College of Medicine, The University of Arizona, Tucson, Arizona, United States
- Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
19
|
Waigi EW, Pernomian L, Crockett AM, Costa TJ, Townsend P, Webb RC, McQuail JA, McCarthy CG, Hollis F, Wenceslau CF. Vascular dysfunction occurs prior to the onset of amyloid pathology and Aβ plaque deposits colocalize with endothelial cells in the hippocampus of female APPswe/PSEN1dE9 mice. GeroScience 2024; 46:5517-5536. [PMID: 38862757 PMCID: PMC11493946 DOI: 10.1007/s11357-024-01213-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/18/2024] [Indexed: 06/13/2024] Open
Abstract
Increasing evidence shows that cardiovascular diseases (CVDs) are associated with an increased risk of cognitive impairment and Alzheimer's diseases (AD). It is unknown whether systemic vascular dysfunction occurs prior to the development of AD, if this occurs in a sex-dependent manner, and whether endothelial cells play a role in the deposition of amyloid beta (Aβ) peptides. We hypothesized that vascular dysfunction occurs prior to the onset of amyloid pathology, thus escalating its progression. Furthermore, endothelial cells from female mice will present with an exacerbated formation of Aβ peptides due to an exacerbated pressure pulsatility. To test this hypothesis, we used a double transgenic mouse model of early-onset AD (APPswe/PSEN1dE9). We evaluated hippocampus-dependent recognition memory and the cardiovascular function by echocardiography and direct measurements of blood pressure through carotid artery catheterization. Vascular function was evaluated in resistance arteries, morphometric parameters in the aortas, and immunofluorescence in the hippocampus and aortas. We observed that endothelial dysfunction occurred prior to the onset of amyloid pathology irrespective of sex. However, during the onset of amyloid pathology, only female APP/PS1 mice had vascular stiffness in the aorta. There was elevated Aβ deposition which colocalized with endothelial cells in the hippocampus from female APP/PS1 mice. Overall, these data showed that vascular abnormalities may be an early marker, and potential mediator of AD, but exacerbated aortic stiffness and pressure pulsatility after the onset of amyloid pathology may be associated with a greater burden of Aβ formation in hippocampal endothelial cells from female but not male APP/PS1 mice.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Laena Pernomian
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Alexia M Crockett
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Tiago J Costa
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Paul Townsend
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, USA
| | - Joseph A McQuail
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, USA
| | - Fiona Hollis
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
20
|
Fontes MT, Costa TJ, de Paula RB, Araújo FA, Barros PR, Townsend P, Butler L, Velazquez KT, Hollis F, Bomfim GF, Butcher JT, McCarthy CG, Wenceslau CF. A Skeletal Muscle-Mediated Anticontractile Response on Vascular Tone: Unraveling the Lactate-AMPK-NOS1 Pathway in Femoral Arteries. FUNCTION 2024; 5:zqae042. [PMID: 39289003 DOI: 10.1093/function/zqae042] [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: 06/19/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
The regulation of vascular tone by perivascular tissues is a complex interplay of various paracrine factors. Here, we investigate the anti-contractile effect of skeletal muscle surrounding the femoral and carotid arteries and its underlying mechanisms. Using male and female Wistar rats, we demonstrated that serotonin, phenylephrine, and U-46619 induced a concentration-dependent vasoconstrictor response in femoral artery rings. Interestingly, this response was diminished in the presence of surrounding femoral skeletal muscle, irrespective of sex. No anti-contractile effect was observed when the carotid artery was exposed to its surrounding skeletal muscle. The observed effect in the femoral artery persisted even in the absence of endothelium and when the muscle was detached from the artery. Furthermore, the skeletal muscle surrounding the femoral artery was able to promote an anti-contractile effect in three other vascular beds (basilar, mesenteric, and carotid arteries). Using inhibitors of lactate dehydrogenase and the 1/4 monocarboxylate transporter, we confirmed the involvement of lactate, as both inhibitors were able to abolish the anti-contractile effect. However, lactate did not directly promote vasodilation; rather, it exerted its effect by activating 5' AMP-activated protein kinase (AMPK) and neuronal nitric oxide synthase (NOS1) in the skeletal muscle. Accordingly, Nω-propyl l-arginine, a specific inhibitor of NOS1, prevented the anti-contractile effect, as well as lactate-induced phosphorylation of NOS1 at the stimulatory serine site (1417) in primary skeletal muscle cells. Phosphorylation of NOS1 was reduced in the presence of Bay-3827, a selective AMPK inhibitor. In conclusion, femoral artery-associated skeletal muscle is a potent paracrine and endocrine organ that influences vascular tone in both sexes. Mechanistically, the anti-contractile effect involves muscle fiber type and/or its anatomical location but not the type of artery or its related vascular endothelium. Finally, the femoral artery anti-contractile effect is mediated by the lactate-AMPK-phospho-NOS1Ser1417-NO signaling axis.
Collapse
Affiliation(s)
- Milene T Fontes
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
| | - Tiago J Costa
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
- Department of Pharmacology, Institute of Biomedical Science, University Of Sao Paulo, Sao Paulo (SP) 05508, Brazil
| | - Ricardo B de Paula
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
| | - Fênix A Araújo
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
| | - Paula R Barros
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
| | - Paul Townsend
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
| | - Landon Butler
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater (OK) 74078, USA
| | - Kandy T Velazquez
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia (SC) 29209, USA
- Columbia Department of Veterans Affairs Health Care System, Columbia (SC) 29209, USA
| | - Fiona Hollis
- Columbia Department of Veterans Affairs Health Care System, Columbia (SC) 29209, USA
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia (SC) 29209, USA
| | - Gisele F Bomfim
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
- Health Research and Education Center, Federal University of Mato Grosso, Sinop (MT) 78556, Brazil
| | - Joshua T Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater (OK) 74078, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
- Biomedical Engineering Program, University of South Carolina, Columbia (SC) 29208, USA
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia (SC) 29209, USA
- Biomedical Engineering Program, University of South Carolina, Columbia (SC) 29208, USA
| |
Collapse
|
21
|
Mastoor Y, Karimi M, Sun M, Ahadi F, Mathieu P, Fan M, Han L, Han LH, Clyne AM. Vascular smooth muscle cells can be circumferentially aligned inside a channel using tunable gelatin microribbons. Biofabrication 2024; 17:015011. [PMID: 39423834 PMCID: PMC11583946 DOI: 10.1088/1758-5090/ad88a7] [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: 04/19/2024] [Accepted: 10/18/2024] [Indexed: 10/21/2024]
Abstract
The gold standard to measure arterial health is vasodilation in response to nitric oxide. Vasodilation is generally measured via pressure myography of arteries isolated from animal models. However, animal arteries can be difficult to obtain and may have limited relevance to human physiology. It is, therefore, critical to engineer human cell-based arterial models capable of contraction. Vascular smooth muscle cells (SMCs) must be circumferentially aligned around the vessel lumen to contract the vessel, which is challenging to achieve in a soft blood vessel model. In this study, we used gelatin microribbons to circumferentially align SMCs inside a hydrogel channel. To accomplish this, we created tunable gelatin microribbons of varying stiffnesses and thicknesses and assessed how SMCs aligned along them. We then wrapped soft, thick microribbons around a needle and encapsulated them in a gelatin methacryloyl hydrogel, forming a microribbon-lined channel. Finally, we seeded SMCs inside the channel and showed that they adhered best to fibronectin and circumferentially aligned in response to the microribbons. Together, these data show that tunable gelatin microribbons can be used to circumferentially align SMCs inside a channel. This technique can be used to create a human artery-on-a-chip to assess vasodilation via pressure myography, as well as to align other cell types for 3Din vitromodels.
Collapse
Affiliation(s)
- Yusuf Mastoor
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, United States of America
| | - Mahsa Karimi
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut St, Philadelphia, PA 19104, United States of America
| | - Michael Sun
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, United States of America
| | - Fereshteh Ahadi
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut St, Philadelphia, PA 19104, United States of America
| | - Pattie Mathieu
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, United States of America
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut St, Philadelphia, PA 19104, United States of America
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut St, Philadelphia, PA 19104, United States of America
| | - Li-Hsin Han
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut St, Philadelphia, PA 19104, United States of America
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, United States of America
| |
Collapse
|
22
|
Haghbin N, Richter DM, Kharche S, Kim MSM, Welsh DG. Functional bias of contractile control in mouse resistance arteries. Sci Rep 2024; 14:24940. [PMID: 39438518 PMCID: PMC11496727 DOI: 10.1038/s41598-024-75838-8] [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: 07/15/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Constrictor agonists set arterial tone through two coupling processes, one tied to (electromechanical), the other independent (pharmacomechanical) of, membrane potential (VM). This dual arrangement raises an intriguing question: is the contribution of each mechanism (1) fixed and proportionate, or (2) variable and functionally biased. Examination began in mouse mesenteric arteries with a vasomotor assessment to a classic Gq/11 (phenylephrine) or Gq/11/G12/13 (U46619) agonist, in the absence and presence of nifedipine, to separate among the two coupling mechanisms. Each constrictor elicited a concentration response curve that was attenuated and rightward shifted by nifedipine, findings consistent with functional bias. Electromechanical coupling preceded pharmacomechanical, the latter's importance rising with agonist concentration. In this regard, ensuing contractile and phosphorylation (CPI-17 & MYPT1 (T-855 & T-697)) measures revealed phenylephrine-induced pharmacomechanical coupling was tied to protein kinase C (PKC) activity, while that enabled by U46619 to PKC and Rho-kinase. A complete switch to pharmacomechanical coupling arose when agonist superfusion was replaced by pipet application to a small portion of artery. This switch was predicted, a priori, by a computer model of electromechanical control and supported by additional measures of VM and cytosolic Ca2+. We conclude that the coupling mechanisms driving agonist-induced constriction are variable and functionally biased, their relative importance set in accordance with agonist concentration and manner of application. These findings have important implications to hemodynamic control in health and disease, including hypertension and arterial vasospasm.
Collapse
Affiliation(s)
- Nadia Haghbin
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - David M Richter
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Sanjay Kharche
- Department of Medical Biophysics, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michelle S M Kim
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Donald G Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
23
|
Islam RA, Han X, Shaligram S, Esfandiarei M, Stallone JN, Rahimian R. Sexual Dimorphism in Impairment of Acetylcholine-Mediated Vasorelaxation in Zucker Diabetic Fatty (ZDF) Rat Aorta: A Monogenic Model of Obesity-Induced Type 2 Diabetes. Int J Mol Sci 2024; 25:11328. [PMID: 39457110 PMCID: PMC11508232 DOI: 10.3390/ijms252011328] [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: 09/10/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Several reports, including our previous studies, indicate that hyperglycemia and diabetes mellitus exert differential effects on vascular function in males and females. This study examines sex differences in the vascular effects of type 2 diabetes (T2D) in an established monogenic model of obesity-induced T2D, Zucker Diabetic Fatty (ZDF) rats. Acetylcholine (ACh) responses were assessed in phenylephrine pre-contracted rings before and after apocynin, a NADPH oxidase (NOX) inhibitor. The mRNA expressions of aortic endothelial NOS (eNOS), and key NOX isoforms were also measured. We demonstrated the following: (1) diabetes had contrasting effects on aortic vasorelaxation in ZDF rats, impairing relaxation to ACh in females while enhancing it in male ZDF rats; (2) inhibition of NOX, a major source of superoxide in vasculature, restored aortic vasorelaxation in female ZDF rats; and (3) eNOS and NOX4 mRNA expressions were elevated in female (but not male) ZDF rat aortas compared to their respective leans. This study highlights sexual dimorphism in ACh-mediated vasorelaxation in the aorta of ZDF rats, suggesting that superoxide may play a role in the impaired vasorelaxation observed in female ZDF rats.
Collapse
Affiliation(s)
- Rifat Ara Islam
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA; (R.A.I.); (S.S.)
| | - Xiaoyuan Han
- Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, Stockton, CA 94115, USA;
| | - Sonali Shaligram
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA; (R.A.I.); (S.S.)
| | - Mitra Esfandiarei
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - John N. Stallone
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute for Comparative Cardiovascular Sciences, School of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4466, USA;
| | - Roshanak Rahimian
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA; (R.A.I.); (S.S.)
| |
Collapse
|
24
|
Munteanu C, Popescu C, Vlădulescu-Trandafir AI, Onose G. Signaling Paradigms of H 2S-Induced Vasodilation: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1158. [PMID: 39456412 PMCID: PMC11505308 DOI: 10.3390/antiox13101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Hydrogen sulfide (H2S), a gas traditionally considered toxic, is now recognized as a vital endogenous signaling molecule with a complex physiology. This comprehensive study encompasses a systematic literature review that explores the intricate mechanisms underlying H2S-induced vasodilation. The vasodilatory effects of H2S are primarily mediated by activating ATP-sensitive potassium (K_ATP) channels, leading to membrane hyperpolarization and subsequent relaxation of vascular smooth muscle cells (VSMCs). Additionally, H2S inhibits L-type calcium channels, reducing calcium influx and diminishing VSMC contraction. Beyond ion channel modulation, H2S profoundly impacts cyclic nucleotide signaling pathways. It stimulates soluble guanylyl cyclase (sGC), increasing the production of cyclic guanosine monophosphate (cGMP). Elevated cGMP levels activate protein kinase G (PKG), which phosphorylates downstream targets like vasodilator-stimulated phosphoprotein (VASP) and promotes smooth muscle relaxation. The synergy between H2S and nitric oxide (NO) signaling further amplifies vasodilation. H2S enhances NO bioavailability by inhibiting its degradation and stimulating endothelial nitric oxide synthase (eNOS) activity, increasing cGMP levels and potent vasodilatory responses. Protein sulfhydration, a post-translational modification, plays a crucial role in cell signaling. H2S S-sulfurates oxidized cysteine residues, while polysulfides (H2Sn) are responsible for S-sulfurating reduced cysteine residues. Sulfhydration of key proteins like K_ATP channels and sGC enhances their activity, contributing to the overall vasodilatory effect. Furthermore, H2S interaction with endothelium-derived hyperpolarizing factor (EDHF) pathways adds another layer to its vasodilatory mechanism. By enhancing EDHF activity, H2S facilitates the hyperpolarization and relaxation of VSMCs through gap junctions between endothelial cells and VSMCs. Recent findings suggest that H2S can also modulate transient receptor potential (TRP) channels, particularly TRPV4 channels, in endothelial cells. Activating these channels by H2S promotes calcium entry, stimulating the production of vasodilatory agents like NO and prostacyclin, thereby regulating vascular tone. The comprehensive understanding of H2S-induced vasodilation mechanisms highlights its therapeutic potential. The multifaceted approach of H2S in modulating vascular tone presents a promising strategy for developing novel treatments for hypertension, ischemic conditions, and other vascular disorders. The interaction of H2S with ion channels, cyclic nucleotide signaling, NO pathways, ROS (Reactive Oxygen Species) scavenging, protein sulfhydration, and EDHF underscores its complexity and therapeutic relevance. In conclusion, the intricate signaling paradigms of H2S-induced vasodilation offer valuable insights into its physiological role and therapeutic potential, promising innovative approaches for managing various vascular diseases through the modulation of vascular tone.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
| | - Cristina Popescu
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Andreea-Iulia Vlădulescu-Trandafir
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
25
|
Yilmaz G, Alexander JS. Impaired Peripheral Vascular Function Following Ischemic Stroke in Mice: Potential Insights into Blood Pressure Variations in the Post-Stroke Patient. PATHOPHYSIOLOGY 2024; 31:488-501. [PMID: 39311310 PMCID: PMC11417821 DOI: 10.3390/pathophysiology31030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
High systolic blood pressure and increased blood pressure variability after the onset of ischemic stroke are associated with poor clinical outcomes. One of the key determinants of blood pressure is arteriolar size, determined by vascular smooth muscle tone and vasodilatory and vasoconstrictor substances that are released by the endothelium. The aim of this study is to outline alterations in vasomotor function in isolated peripheral arteries following ischemic stroke. The reactivity of thoracic aortic segments from male C57BL/6 mice to dilators and constrictors was quantified using wire myography. Acetylcholine-induced endothelium-dependent vasodilation was impaired after ischemic stroke (LogIC50 Sham = -7.499, LogIC50 Stroke = -7.350, p = 0.0132, n = 19, 31 respectively). The vasodilatory responses to SNP were identical in the isolated aortas in the sham and stroke groups. Phenylephrine-induced vasoconstriction was impaired in the aortas isolated from the stroke animals in comparison to their sham treatment counterparts (Sham LogEC50= -6.652 vs. Stroke LogEC50 = -6.475, p < 0.001). Our study demonstrates that 24 h post-ischemic stroke, peripheral vascular responses are impaired in remote arteries. The aortas from the stroke animals exhibited reduced vasoconstrictor and endothelium-dependent vasodilator responses, while the endothelium-independent vasodilatory responses were preserved. Since both the vasodilatory and vasoconstrictor responses of peripheral arteries are impaired following ischemic stroke, our findings might explain increased blood pressure variability following ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Molecular Cellular and Biomedical Sciences, CUNY School of Medicine, New York, NY 10031, USA
| | - Jonathan Steven Alexander
- Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA;
| |
Collapse
|
26
|
Fato BR, de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu'u-Lino TJ, Bubb KJ, Hannan NJ. Exploring the Therapeutic Potential of C-Type Natriuretic Peptide for Preeclampsia. Hypertension 2024; 81:1883-1894. [PMID: 39016006 DOI: 10.1161/hypertensionaha.124.22820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Preeclampsia is a serious condition of pregnancy, complicated by aberrant maternal vascular dysfunction. CNP (C-type natriuretic peptide) contributes to vascular homeostasis, acting through NPR-B (natriuretic peptide receptor-B) and NPR-C (natriuretic peptide receptor-C). CNP mitigates vascular dysfunction of arteries in nonpregnant cohorts; this study investigates whether CNP can dilate maternal arteries in ex vivo preeclampsia models. METHODS Human omental arteries were dissected from fat biopsies collected during cesarean section. CNP, NPR-B, and NPR-C mRNA expression was assessed in arteries collected from pregnancies complicated by preeclampsia (n=6) and normotensive controls (n=11). Using wire myography, we investigated the effects of CNP on dilation of arteries from normotensive pregnancies. Arteries were preconstricted with either serum from patients with preeclampsia (n=6) or recombinant ET-1 (endothelin-1; vasoconstrictor elevated in preeclampsia; n=6) to model vasoconstriction associated with preeclampsia. Preconstricted arteries were treated with recombinant CNP (0.001-100 µmol/L) or vehicle and vascular relaxation assessed. In further studies, arteries were preincubated with NPR-B (5 µmol/L) and NPR-C (10 µmol/L) antagonists before serum-induced constriction (n=4-5) to explore mechanistic signaling. RESULTS CNP, NPR-B, and NPR-C mRNAs were not differentially expressed in omental arteries from preeclamptic pregnancies. CNP potently stimulated maternal artery vasorelaxation in our model of preeclampsia (using preeclamptic serum). Its vasodilatory actions were driven through the activation of NPR-B predominantly; antagonism of this receptor alone dampened CNP vasorelaxation. Interestingly, CNP did not reduce ET-1-driven omental artery constriction. CONCLUSIONS Collectively, these data suggest that enhancing CNP signaling through NPR-B offers a potential therapeutic strategy to reduce systemic vascular constriction in preeclampsia.
Collapse
Affiliation(s)
- Bianca R Fato
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natalie K Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Kristen J Bubb
- Department of Physiology, Biomedicine Discovery Institute (K.J.B.), Monash University, Clayton, Victoria, Australia
- Victorian Heart Institute, Faculty of Medicine, Nursing and Health Sciences (K.J.B.), Monash University, Clayton, Victoria, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
27
|
Myers TD, Li Y, Taiclet S, Cabada-Aguirre P, Kuti E, McClure K, Blanchard C, Wolosowicz M, Homanics GE, Straub AC, Meriney SD, Palladino MJ. Neuromuscular dysfunction and pathogenesis in triosephosphate isomerase deficiency. Sci Rep 2024; 14:18575. [PMID: 39127839 PMCID: PMC11316810 DOI: 10.1038/s41598-024-69618-7] [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: 06/07/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024] Open
Abstract
Triosephosphate isomerase deficiency (TPI Df) is a rare multisystem disorder with severe neuromuscular symptoms which arises exclusively from mutations within the TPI1 gene. Studies of TPI Df have been limited due to the absence of mammalian disease models and difficulties obtaining patient samples. Recently, we developed a novel murine model of TPI Df which models the most common disease-causing mutation in humans, TPI1E105D. Using our model in the present study, the underlying pathogenesis of neuromuscular symptoms has been elucidated. This is the first report detailing studies of neuromuscular pathology within a murine model of TPI Df. We identified several contributors to neuromuscular symptoms, including neurodegeneration in the brain, alterations in neurotransmission at the neuromuscular junction, and reduced muscle fiber size. TPI Df mice also exhibited signs of cardiac pathology and displayed a deficit in vascular smooth muscle functionality. Together, these findings provide insight into pathogenesis of the neuromuscular symptoms in TPI Df and can guide the future development of therapeutics.
Collapse
Affiliation(s)
- Tracey D Myers
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yizhi Li
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stefanie Taiclet
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paulina Cabada-Aguirre
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily Kuti
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaitlin McClure
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Marta Wolosowicz
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gregg E Homanics
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Preoperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen D Meriney
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Palladino
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Arsyad A, Lembang GKR, Linda SL, Djabir YY, Dobson GP. Low Calcium-High Magnesium Krebs-Henseleit Solution Combined with Adenosine and Lidocaine Improved Rat Aortic Function and Structure Following Cold Preservation. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1284. [PMID: 39202566 PMCID: PMC11356418 DOI: 10.3390/medicina60081284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024]
Abstract
Background and objectives: The main problem of vascular preservation is the maintenance of vessel graft quality and function following extended storage. Conventional preservation solutions such as histidine-tryptophan-ketoglutarate (HTK) solution, Phosphate-Buffer Solution (PBS), or sodium chloride 0.9% has been shown to be inadequate in preserving vascular physiological function after 3 days of cold storage. This study aimed to evaluate whether adenosine and lidocaine (AL) in a modified Krebs-Henseleit (KH) solution can preserve the function and histological structure of rat aortic rings after 6 days. Materials and Methods: Thirty-five aortic rings from male Wistar rats (200-300 g) were harvested and immediately immersed in one of the assigned cold preservation solutions: standard KH, modified KH (mod KH) with lower calcium (Ca2+) and higher magnesium content (Mg2+) with or without adenosine and lidocaine (mod KH-AL), and modified KH with AL, insulin, and melatonin (Mod KH-ALMI). The contraction and relaxation function of the aortic rings were examined using an isometric force transducer after 6 days of cold preservation. Hematoxylin and eosin staining were used to analyze the rings' histological structure. Results: Vascular contraction and relaxation functions were severely affected after a 6-day cold storage period in standard KH. Modifying the KH solution by reducing the Ca2+ and increasing the Mg2+ levels greatly recovered the vessel functions. The addition of AL or ALMI to the modified KH did not further recover vascular contractility. However, only the addition of AL to the modified KH increased the ACh-induced relaxation at 6 days when compared to the conventional KH, suggesting that endothelium preservation is improved. From histological analysis, it was found that the addition of AL but not ALMI further improved the endothelial lining and the structure of the elastic membrane layers of the preserved vessels after 6 days of cold preservation. Conclusions: The addition of AL to low calcium-high magnesium KH solution significantly enhanced endothelial preservation and improved endothelial-induced relaxation of preserved vessels after 6 days of cold storage.
Collapse
Affiliation(s)
- Aryadi Arsyad
- Department of Physiology, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Geni K. R. Lembang
- Clinical Pharmacy Laboratory, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (G.K.R.L.); (S.L.L.); (Y.Y.D.)
| | - Sesilia L. Linda
- Clinical Pharmacy Laboratory, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (G.K.R.L.); (S.L.L.); (Y.Y.D.)
| | - Yulia Y. Djabir
- Clinical Pharmacy Laboratory, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (G.K.R.L.); (S.L.L.); (Y.Y.D.)
| | - Geoffrey P. Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia;
| |
Collapse
|
29
|
Metwally E, Sanchez Solano A, Lavanderos B, Yamasaki E, Thakore P, McClenaghan C, Rios N, Radi R, Feng Earley Y, Nichols CG, Earley S. Mitochondrial Ca2+-coupled generation of reactive oxygen species, peroxynitrite formation, and endothelial dysfunction in Cantú syndrome. JCI Insight 2024; 9:e176212. [PMID: 39088268 PMCID: PMC11385080 DOI: 10.1172/jci.insight.176212] [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: 09/27/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Cantú syndrome is a multisystem disorder caused by gain-of-function (GOF) mutations in KCNJ8 and ABCC9, the genes encoding the pore-forming inward rectifier Kir6.1 and regulatory sulfonylurea receptor SUR2B subunits, respectively, of vascular ATP-sensitive K+ (KATP) channels. In this study, we investigated changes in the vascular endothelium in mice in which Cantú syndrome-associated Kcnj8 or Abcc9 mutations were knocked in to the endogenous loci. We found that endothelium-dependent dilation was impaired in small mesenteric arteries from Cantú mice. Loss of endothelium-dependent vasodilation led to increased vasoconstriction in response to intraluminal pressure or treatment with the adrenergic receptor agonist phenylephrine. We also found that either KATP GOF or acute activation of KATP channels with pinacidil increased the amplitude and frequency of wave-like Ca2+ events generated in the endothelium in response to the vasodilator agonist carbachol. Increased cytosolic Ca2+ signaling activity in arterial endothelial cells from Cantú mice was associated with elevated mitochondrial [Ca2+] and enhanced reactive oxygen species (ROS) and peroxynitrite levels. Scavenging intracellular or mitochondrial ROS restored endothelium-dependent vasodilation in the arteries of mice with KATP GOF mutations. We conclude that mitochondrial Ca2+ overload and ROS generation, which subsequently leads to nitric oxide consumption and peroxynitrite formation, cause endothelial dysfunction in mice with Cantú syndrome.
Collapse
Affiliation(s)
- Elsayed Metwally
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Boris Lavanderos
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Conor McClenaghan
- Departments of Pharmacology and Medicine, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Natalia Rios
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Yumei Feng Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Colin G. Nichols
- Center for the Investigation of Membrane Excitability Diseases and Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| |
Collapse
|
30
|
de Vries T, Labruijere S, Rivera-Mancilla E, Garrelds IM, de Vries R, Schutter D, van den Bogaerdt A, Poyner DR, Ladds G, Danser AHJ, MaassenVanDenBrink A. Intracellular pathways of calcitonin gene-related peptide-induced relaxation of human coronary arteries: A key role for Gβγ subunit instead of cAMP. Br J Pharmacol 2024; 181:2478-2491. [PMID: 38583945 DOI: 10.1111/bph.16372] [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: 11/15/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND AND PURPOSE Calcitonin gene-related peptide (CGRP) is a potent vasodilator. While its signalling is assumed to be mediated via increases in cAMP, this study focused on elucidating the actual intracellular signalling pathways involved in CGRP-induced relaxation of human isolated coronary arteries (HCA). EXPERIMENTAL APPROACH HCA were obtained from heart valve donors (27 M, 25 F, age 54 ± 2 years). Concentration-response curves to human α-CGRP or forskolin were constructed in HCA segments, incubated with different inhibitors of intracellular signalling pathways, and intracellular cAMP levels were measured with and without stimulation. RESULTS Adenylyl cyclase (AC) inhibitors SQ22536 + DDA and MDL-12330A, and PKA inhibitors Rp-8-Br-cAMPs and H89, did not inhibit CGRP-induced relaxation of HCA, nor did the guanylyl cyclase inhibitor ODQ, PKG inhibitor KT5823, EPAC1/2 inhibitor ESI09, potassium channel blockers TRAM-34 + apamin, iberiotoxin or glibenclamide, or the Gαq inhibitor YM-254890. Phosphodiesterase inhibitors induced a concentration-dependent decrease in the response to KCl but did not potentiate relaxation to CGRP. Relaxation to forskolin was not blocked by PKA or AC inhibitors, although AC inhibitors significantly inhibited the increase in cAMP. Inhibition of Gβγ subunits using gallein significantly inhibited the relaxation to CGRP in human coronary arteries. CONCLUSION While CGRP signalling is generally assumed to act via cAMP, the CGRP-induced vasodilation in HCA was not inhibited by targeting this intracellular signalling pathway at different levels. Instead, inhibition of Gβγ subunits did inhibit the relaxation to CGRP, suggesting a different mechanism of CGRP-induced relaxation than generally believed.
Collapse
Affiliation(s)
- Tessa de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sieneke Labruijere
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - René de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Dennis Schutter
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - David R Poyner
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Singhrao N, Flores-Tamez VA, Moustafa YA, Reddy GR, Burns AE, Pinkerton KE, Chen CY, Navedo MF, Nieves-Cintrón M. Nicotine Impairs Smooth Muscle cAMP Signaling and Vascular Reactivity. Microcirculation 2024; 31:e12871. [PMID: 38805589 PMCID: PMC11303104 DOI: 10.1111/micc.12871] [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: 11/14/2023] [Revised: 03/29/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
OBJECTIVE This study aimed to determine nicotine's impact on receptor-mediated cyclic adenosine monophosphate (cAMP) synthesis in vascular smooth muscle (VSM). We hypothesize that nicotine impairs β adrenergic-mediated cAMP signaling in VSM, leading to altered vascular reactivity. METHODS The effects of nicotine on cAMP signaling and vascular function were systematically tested in aortic VSM cells and acutely isolated aortas from mice expressing the cAMP sensor TEpacVV (Camper), specifically in VSM (e.g., CamperSM). RESULTS Isoproterenol (ISO)-induced β-adrenergic production of cAMP in VSM was significantly reduced in cells from second-hand smoke (SHS)-exposed mice and cultured wild-type VSM treated with nicotine. The decrease in cAMP synthesis caused by nicotine was verified in freshly isolated arteries from a mouse that had cAMP sensor expression in VSM (e.g., CamperSM mouse). Functionally, the changes in cAMP signaling in response to nicotine hindered ISO-induced vasodilation, but this was reversed by immediate PDE3 inhibition. CONCLUSIONS These results imply that nicotine alters VSM β adrenergic-mediated cAMP signaling and vasodilation, which may contribute to the dysregulation of vascular reactivity and the development of vascular complications for nicotine-containing product users.
Collapse
Affiliation(s)
- Navid Singhrao
- Department of Pharmacology, University of California, Davis, USA
| | | | | | | | - Abby E. Burns
- Department of Pharmacology, University of California, Davis, USA
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, California, USA
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, USA
| | | |
Collapse
|
32
|
Colebank MJ, Oomen PA, Witzenburg CM, Grosberg A, Beard DA, Husmeier D, Olufsen MS, Chesler NC. Guidelines for mechanistic modeling and analysis in cardiovascular research. Am J Physiol Heart Circ Physiol 2024; 327:H473-H503. [PMID: 38904851 PMCID: PMC11442102 DOI: 10.1152/ajpheart.00766.2023] [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: 12/11/2023] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Computational, or in silico, models are an effective, noninvasive tool for investigating cardiovascular function. These models can be used in the analysis of experimental and clinical data to identify possible mechanisms of (ab)normal cardiovascular physiology. Recent advances in computing power and data management have led to innovative and complex modeling frameworks that simulate cardiovascular function across multiple scales. While commonly used in multiple disciplines, there is a lack of concise guidelines for the implementation of computer models in cardiovascular research. In line with recent calls for more reproducible research, it is imperative that scientists adhere to credible practices when developing and applying computational models to their research. The goal of this manuscript is to provide a consensus document that identifies best practices for in silico computational modeling in cardiovascular research. These guidelines provide the necessary methods for mechanistic model development, model analysis, and formal model calibration using fundamentals from statistics. We outline rigorous practices for computational, mechanistic modeling in cardiovascular research and discuss its synergistic value to experimental and clinical data.
Collapse
Affiliation(s)
- Mitchel J Colebank
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States
| | - Pim A Oomen
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States
| | - Colleen M Witzenburg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Anna Grosberg
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Dirk Husmeier
- School of Mathematics and Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Mette S Olufsen
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, United States
| | - Naomi C Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States
| |
Collapse
|
33
|
Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci (Lond) 2024; 138:817-850. [PMID: 38920058 DOI: 10.1042/cs20220797] [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: 09/26/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1β and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-β/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.
Collapse
Affiliation(s)
- Justyna Totoń-Żurańska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Blessy Saju
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| | - Tomasz J Guzik
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
34
|
Collins HE, Alexander BT, Care AS, Davenport MH, Davidge ST, Eghbali M, Giussani DA, Hoes MF, Julian CG, LaVoie HA, Olfert IM, Ozanne SE, Bytautiene Prewit E, Warrington JP, Zhang L, Goulopoulou S. Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum. Am J Physiol Heart Circ Physiol 2024; 327:H191-H220. [PMID: 38758127 PMCID: PMC11380979 DOI: 10.1152/ajpheart.00055.2024] [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: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.
Collapse
Grants
- HL169157 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HD088590 NICHD NIH HHS
- HD083132 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- The Biotechnology and Biological Sciences Research Council
- P20GM103499 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- British Heart Foundation (BHF)
- R21 HD111908 NICHD NIH HHS
- Distinguished University Professor
- The Lister Insititute
- ES032920 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL149608 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Royal Society (The Royal Society)
- U.S. Department of Defense (DOD)
- HL138181 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- MC_00014/4 UKRI | Medical Research Council (MRC)
- RG/17/8/32924 British Heart Foundation
- Jewish Heritage Fund for Excellence
- HD111908 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP2002129 NHMRC Ideas Grant
- HL159865 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL131182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NS103017 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL138181 NHLBI NIH HHS
- 20CSA35320107 American Heart Association (AHA)
- RG/17/12/33167 British Heart Foundation (BHF)
- National Heart Foundation Future Leader Fellowship
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155295 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HD088590-06 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- WVU SOM Synergy Grant
- R01 HL146562 NHLBI NIH HHS
- R01 HL159865 NHLBI NIH HHS
- Canadian Insitute's of Health Research Foundation Grant
- R01 HL169157 NHLBI NIH HHS
- HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ES034646-01 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL150472 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 2021T017 Dutch Heart Foundation Dekker Grant
- MC_UU_00014/4 Medical Research Council
- R01 HL163003 NHLBI NIH HHS
- Christenson professor In Active Healthy Living
- National Heart Foundation
- Dutch Heart Foundation Dekker
- WVU SOM Synergy
- Jewish Heritage
- Department of Health | National Health and Medical Research Council (NHMRC)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Helen E Collins
- University of Louisville, Louisville, Kentucky, United States
| | - Barbara T Alexander
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alison S Care
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | - Mansoureh Eghbali
- University of California Los Angeles, Los Angeles, California, United States
| | | | | | - Colleen G Julian
- University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Holly A LaVoie
- University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - I Mark Olfert
- West Virginia University School of Medicine, Morgantown, West Virginia, United States
| | | | | | - Junie P Warrington
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Lubo Zhang
- Loma Linda University School of Medicine, Loma Linda, California, United States
| | | |
Collapse
|
35
|
Warrington JP, Collins HE, Davidge ST, do Carmo JM, Goulopoulou S, Intapad S, Loria AS, Sones JL, Wold LE, Zinkhan EK, Alexander BT. Guidelines for in vivo models of developmental programming of cardiovascular disease risk. Am J Physiol Heart Circ Physiol 2024; 327:H221-H241. [PMID: 38819382 PMCID: PMC11380980 DOI: 10.1152/ajpheart.00060.2024] [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: 01/31/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Research using animals depends on the generation of offspring for use in experiments or for the maintenance of animal colonies. Although not considered by all, several different factors preceding and during pregnancy, as well as during lactation, can program various characteristics in the offspring. Here, we present the most common models of developmental programming of cardiovascular outcomes, important considerations for study design, and provide guidelines for producing and reporting rigorous and reproducible cardiovascular studies in offspring exposed to normal conditions or developmental insult. These guidelines provide considerations for the selection of the appropriate animal model and factors that should be reported to increase rigor and reproducibility while ensuring transparent reporting of methods and results.
Collapse
Grants
- 20YVNR35490079 American Heart Association (AHA)
- R01HL139348 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135158 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54GM115428 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01AG057046 HHS | NIH | National Institute on Aging (NIA)
- P20 GM104357 NIGMS NIH HHS
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 GM149404 NIGMS NIH HHS
- P20GM104357 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM135002 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL163003 NHLBI NIH HHS
- R01HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01DK121411 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Excellence Faculty Support Grant Jewish Heritage Fund
- P30GM149404 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P30GM14940 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- 23SFRNPCS1067044 American Heart Association (AHA)
- R01 HL146562 NHLBI NIH HHS
- R56HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54 GM115428 NIGMS NIH HHS
- 1R01HL163076 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL51971 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- FS154313 CIHR
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Helen E Collins
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, Kentucky, United States
| | - Sandra T Davidge
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jussara M do Carmo
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, United States
- Department of Gynecology, and Obstetrics, Loma Linda University, Loma Linda, California, United States
| | - Suttira Intapad
- Department of Pharmacology, Tulane University, New Orleans, Louisiana, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jenny L Sones
- Equine Reproduction Laboratory, Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Erin K Zinkhan
- Department of Pediatrics, University of Utah and Intermountain Health, Salt Lake City, Utah, United States
- Intermountain Health, Salt Lake City, Utah, United States
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
36
|
Cevik E, Albadawi H, Zhang Z, Demirlenk Y, Atar D, Keum C, Kim J, Graf E, Gunduz S, Rehman S, Oklu R. Catheter-Directed Ionic Liquid Embolic Agent for Rapid Portal Vein Embolization, Segmentectomy, and Bile Duct Ablation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402570. [PMID: 38678378 PMCID: PMC11257814 DOI: 10.1002/adma.202402570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/21/2024] [Indexed: 04/29/2024]
Abstract
Embolic materials currently in use for portal vein embolization (PVE) do not treat the tumor, which poses a risk for tumor progression during the interval between PVE and surgical resection. Here, is developed an ionic-liquid-based embolic material (LEAD) for portal vein embolization, liver ablation, and drug delivery. LEAD is optimized and characterized for diffusivity, X-ray visibility, and cytotoxicity. In the porcine renal embolization model, LEAD delivered from the main renal artery reached vasculature down to 10 microns with uniform tissue ablation and delivery of small and large therapeutics. In non-survival and survival porcine experiments, successful PVE is achieved in minutes, leading to the expected chemical segmentectomy, and delivery of a large protein drug (i.e., Nivolumab) with LEAD. In cholangiocarcinoma mouse tumor models and in ex vivo human tumors, LEAD consistently achieved an effective ablation and wide drug distribution. Furthermore, various strains of drug-resistant patient-derived bacteria showed significant susceptibility to LEAD, suggesting that LEAD may also prevent infectious complications resulting from tissue ablation. With its capabilities to embolize, ablate, and deliver therapeutics, ease of use, and a high safety profile demonstrated in animal studies, LEAD offers a potential alternative to tumor ablation with or without PVE for FLR growth.
Collapse
Affiliation(s)
- Enes Cevik
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Hassan Albadawi
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Zefu Zhang
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Yusuf Demirlenk
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Dila Atar
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Chris Keum
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Jinjoo Kim
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Erin Graf
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, Arizona 85054, USA
| | - Seyda Gunduz
- Department of Medical Oncology, Istinye University Bahcesehir Liv Hospital, Istanbul 34517, Turkey
| | - Suliman Rehman
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Rahmi Oklu
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| |
Collapse
|
37
|
Trotta RJ, Harmon DL, Klotz JL. Serotonin receptor-mediated vasorelaxation occurs primarily through 5-HT 4 activation in bovine lateral saphenous vein. Physiol Rep 2024; 12:e16128. [PMID: 38946059 PMCID: PMC11214916 DOI: 10.14814/phy2.16128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 07/02/2024] Open
Abstract
To better understand mechanisms of serotonin- (5-HT) mediated vasorelaxation, isolated lateral saphenous veins from cattle were assessed for vasoactivity using myography in response to increasing concentrations of 5-HT or selective 5-HT receptor agonists. Vessels were pre-contracted with 1 × 10-4 M phenylephrine and exposed to increasing concentrations of 5-HT or 5-HT receptor agonists that were selective for 5-HT1B, 5-HT2B, 5-HT4, and 5-HT7. Vasoactive response data were normalized as a percentage of the maximum contractile response induced by the phenylephrine pre-contraction. At 1 × 10-7 M 5-HT, a relaxation was observed with an 88.7% decrease (p < 0.01) from the phenylephrine maximum. At 1 × 10-4 M 5-HT, a contraction was observed with a 165% increase (p < 0.01) from the phenylephrine maximum. Increasing concentrations of agonists selective for 5-HT2B, 5-HT4, or 5-HT7 resulted in a 27%, 92%, or 44% (p < 0.01) decrease from the phenylephrine maximum, respectively. Of these 5-HT receptor agonists, the selective 5-HT4 receptor agonist resulted in the greatest potency (-log EC50) value (6.30) compared with 5-HT2B and 5-HT7 receptor agonists (4.21 and 4.66, respectively). To confirm the involvement of 5-HT4 in 5-HT-mediated vasorelaxation, blood vessels were exposed to either DMSO (solvent control) or a selective 5-HT4 antagonist (1 × 10-5 M) for 5-min prior to the phenylephrine pre-contraction and 5-HT additions. Antagonism of the 5-HT4 receptor attenuated the vasorelaxation caused by 5-HT. Approximately 94% of the vasorelaxation occurring in response to 5-HT could be accounted for through 5-HT4, providing strong evidence that 5-HT-mediated vasorelaxation occurs through 5-HT4 activation in bovine peripheral vasculature.
Collapse
Affiliation(s)
- Ronald J. Trotta
- Department of Animal and Food SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - David L. Harmon
- Department of Animal and Food SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - James L. Klotz
- Forage‐Animal Production Research UnitUSDA‐ARSLexingtonKentuckyUSA
| |
Collapse
|
38
|
Wahl D, Clayton ZS. Peripheral vascular dysfunction and the aging brain. Aging (Albany NY) 2024; 16:9280-9302. [PMID: 38805248 PMCID: PMC11164523 DOI: 10.18632/aging.205877] [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: 01/04/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024]
Abstract
Aging is the greatest non-modifiable risk factor for most diseases, including cardiovascular diseases (CVD), which remain the leading cause of mortality worldwide. Robust evidence indicates that CVD are a strong determinant for reduced brain health and all-cause dementia with advancing age. CVD are also closely linked with peripheral and cerebral vascular dysfunction, common contributors to the development and progression of all types of dementia, that are largely driven by excessive levels of oxidative stress (e.g., reactive oxygen species [ROS]). Emerging evidence suggests that several fundamental aging mechanisms (e.g., "hallmarks" of aging), including chronic low-grade inflammation, mitochondrial dysfunction, cellular senescence and deregulated nutrient sensing contribute to excessive ROS production and are common to both peripheral and cerebral vascular dysfunction. Therefore, targeting these mechanisms to reduce ROS-related oxidative stress and improve peripheral and/or cerebral vascular function may be a promising strategy to reduce dementia risk with aging. Investigating how certain lifestyle strategies (e.g., aerobic exercise and diet modulation) and/or select pharmacological agents (natural and synthetic) intersect with aging "hallmarks" to promote peripheral and/or cerebral vascular health represent a viable option for reducing dementia risk with aging. Therefore, the primary purpose of this review is to explore mechanistic links among peripheral vascular dysfunction, cerebral vascular dysfunction, and reduced brain health with aging. Such insight and assessments of non-invasive measures of peripheral and cerebral vascular health with aging might provide a new approach for assessing dementia risk in older adults.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science and Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Zachary S. Clayton
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Geriatric Medicine, Aurora, CO 80045, USA
| |
Collapse
|
39
|
Mahoney SA, VanDongen NS, Greenberg NT, Venkatasubramanian R, Rossman MJ, Widlansky ME, Brunt VE, Bernaldo de Quirós Y, Seals DR, Clayton ZS. Role of the circulating milieu in age-related arterial dysfunction: a novel ex vivo approach. Am J Physiol Heart Circ Physiol 2024; 326:H1279-H1290. [PMID: 38517225 PMCID: PMC11380963 DOI: 10.1152/ajpheart.00014.2024] [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: 01/04/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
The circulating milieu, bioactive molecules in the bloodstream, is altered with aging and interfaces constantly with the vasculature. This anatomic juxtaposition suggests that circulating factors may actively modulate arterial function. Here, we developed a novel, translational experimental model that allows for direct interrogation of the influence of the circulating milieu on age-related arterial dysfunction (aortic stiffening and endothelial dysfunction). To do so, we exposed young and old mouse arteries to serum from young and old mice and young and midlife/older (ML/O) adult humans. We found that old mouse and ML/O adult human, but not young, serum stiffened young mouse aortic rings, assessed via elastic modulus (mouse and human serum, P = 0.003 vs. young serum control), and impaired carotid artery endothelial function, assessed by endothelium-dependent dilation (EDD) (mouse serum, P < 0.001; human serum, P = 0.006 vs. young serum control). Furthermore, young mouse and human, but not old, serum reduced aortic elastic modulus (mouse serum, P = 0.009; human serum, P < 0.001 vs. old/MLO serum control) and improved EDD (mouse and human serum, P = 0.015 vs. old/MLO serum control) in old arteries. In human serum-exposed arteries, in vivo arterial function assessed in the human donors correlated with circulating milieu-modulated arterial function in young mouse arteries (aortic stiffness, r = 0.634, P = 0.005; endothelial function, r = 0.609, P = 0.004) and old mouse arteries (aortic stiffness, r = 0.664, P = 0.001; endothelial function, r = 0.637, P = 0.003). This study establishes novel experimental approaches for directly assessing the effects of the circulating milieu on arterial function and implicates changes in the circulating milieu as a mechanism of in vivo arterial aging.NEW & NOTEWORTHY Changes in the circulating milieu with advancing age may be a mechanism underlying age-related arterial dysfunction. Ex vivo exposure of young mouse arteries to the circulating milieu from old mice or midlife/older adults impairs arterial function whereas exposure of old mouse arteries to the circulating milieu from young mice or young adults improves arterial function. These findings establish that the circulating milieu directly influences arterial function with aging.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nicholas S VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Michael E Widlansky
- Department of Medicine and Pharmacology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Yara Bernaldo de Quirós
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
40
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
41
|
Lindsey ML, Kirk JA, LeBlanc AJ, Brunt KR, Carter JR, Hansell Keehan K, Ripplinger CM, Kleinbongard P, Kassiri Z. Looking backward to plan forward. Am J Physiol Heart Circ Physiol 2024; 326:H1155-H1158. [PMID: 38551484 DOI: 10.1152/ajpheart.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Affiliation(s)
- Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois, United States
| | - Amanda J LeBlanc
- Department of Cardiovascular and Thoracic Surgery and Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jason R Carter
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, United States
| | - Kara Hansell Keehan
- American Journal of Physiology-Heart and Circulatory Physiology, American Physiological Society, Rockville, Maryland, United States
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, United States
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
42
|
Fan L, Wang H, Kassab GS, Lee LC. Review of cardiac-coronary interaction and insights from mathematical modeling. WIREs Mech Dis 2024; 16:e1642. [PMID: 38316634 PMCID: PMC11081852 DOI: 10.1002/wsbm.1642] [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: 09/13/2023] [Revised: 12/10/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Cardiac-coronary interaction is fundamental to the function of the heart. As one of the highest metabolic organs in the body, the cardiac oxygen demand is met by blood perfusion through the coronary vasculature. The coronary vasculature is largely embedded within the myocardial tissue which is continually contracting and hence squeezing the blood vessels. The myocardium-coronary vessel interaction is two-ways and complex. Here, we review the different types of cardiac-coronary interactions with a focus on insights gained from mathematical models. Specifically, we will consider the following: (1) myocardial-vessel mechanical interaction; (2) metabolic-flow interaction and regulation; (3) perfusion-contraction matching, and (4) chronic interactions between the myocardium and coronary vasculature. We also provide a discussion of the relevant experimental and clinical studies of different types of cardiac-coronary interactions. Finally, we highlight knowledge gaps, key challenges, and limitations of existing mathematical models along with future research directions to understand the unique myocardium-coronary coupling in the heart. This article is categorized under: Cardiovascular Diseases > Computational Models Cardiovascular Diseases > Biomedical Engineering Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Lei Fan
- Joint Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Haifeng Wang
- Department of Mechanical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ghassan S Kassab
- California Medical Innovations Institute, San Diego, California, USA
| | - Lik Chuan Lee
- Department of Mechanical Engineering, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
43
|
Asunción-Alvarez D, Palacios J, Ybañez-Julca RO, Rodriguez-Silva CN, Nwokocha C, Cifuentes F, Greensmith DJ. Calcium signaling in endothelial and vascular smooth muscle cells: sex differences and the influence of estrogens and androgens. Am J Physiol Heart Circ Physiol 2024; 326:H950-H970. [PMID: 38334967 DOI: 10.1152/ajpheart.00600.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Calcium signaling in vascular endothelial cells (ECs) and smooth muscle cells (VSMCs) is essential for the regulation of vascular tone. However, the changes to intracellular Ca2+ concentrations are often influenced by sex differences. Furthermore, a large body of evidence shows that sex hormone imbalance leads to dysregulation of Ca2+ signaling and this is a key factor in the pathogenesis of cardiovascular diseases. In this review, the effects of estrogens and androgens on vascular calcium-handling proteins are discussed, with emphasis on the associated genomic or nongenomic molecular mechanisms. The experimental models from which data were collected were also considered. The review highlights 1) in female ECs, transient receptor potential vanilloid 4 (TRPV4) and mitochondrial Ca2+ uniporter (MCU) enhance Ca2+-dependent nitric oxide (NO) generation. In males, only transient receptor potential canonical 3 (TRPC3) plays a fundamental role in this effect. 2) Female VSMCs have lower cytosolic Ca2+ levels than males due to differences in the activity and expression of stromal interaction molecule 1 (STIM1), calcium release-activated calcium modulator 1 (Orai1), calcium voltage-gated channel subunit-α1C (CaV1.2), Na+-K+-2Cl- symporter (NKCC1), and the Na+/K+-ATPase. 3) When compared with androgens, the influence of estrogens on Ca2+ homeostasis, vascular tone, and incidence of vascular disease is better documented. 4) Many studies use supraphysiological concentrations of sex hormones, which may limit the physiological relevance of outcomes. 5) Sex-dependent differences in Ca2+ signaling mean both sexes ought to be included in experimental design.
Collapse
Affiliation(s)
- Daniel Asunción-Alvarez
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Javier Palacios
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Roberto O Ybañez-Julca
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Cristhian N Rodriguez-Silva
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences Physiology Section, Faculty of Medical Sciences, The University of the West Indies, Kingston, Jamaica
| | - Fredi Cifuentes
- Laboratorio de Fisiología Experimental (EphyL), Instituto Antofagasta (IA), Universidad de Antofagasta, Antofagasta, Chile
| | - David J Greensmith
- Biomedical Research Centre, School of Science, Engineering and Environment, The University of Salford, Salford, United Kingdom
| |
Collapse
|
44
|
Chu Z, Kassmann M, Anistan YM, Luft FC, Gollasch M, Tsvetkov D. Protocol for assessing myogenic tone and perfusion pressure in isolated mouse kidneys. STAR Protoc 2024; 5:102845. [PMID: 38294910 PMCID: PMC10844887 DOI: 10.1016/j.xpro.2024.102845] [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: 07/24/2023] [Revised: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
The isolated perfused kidney is a classic ex vivo preparation for studying renal physiology in general and vascular function. Here, we present a protocol for assessing myogenic tone in isolated mouse kidneys as well as vasodilatory and vasoconstrictive responses, expressed as perfusion pressure. We describe steps for pre-operative preparation, kidney and renal artery isolation, and connection of renal artery with glass cannula. We then detail how to measure pressure changes in perfused kidneys and the myogenic tone. For complete details on the use and execution of this protocol, please refer to Cui et al.1.
Collapse
Affiliation(s)
- Zhugang Chu
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17489 Greifswald, Germany; Department of Urology, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Mario Kassmann
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Yoland-Marie Anistan
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17489 Greifswald, Germany.
| | - Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, 17489 Greifswald, Germany.
| |
Collapse
|
45
|
Reeve EH, Barnes JN, Moir ME, Walker AE. Impact of arterial stiffness on cerebrovascular function: a review of evidence from humans and preclincal models. Am J Physiol Heart Circ Physiol 2024; 326:H689-H704. [PMID: 38214904 PMCID: PMC11221809 DOI: 10.1152/ajpheart.00592.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
With advancing age, the cerebral vasculature becomes dysfunctional, and this dysfunction is associated with cognitive decline. However, the initiating cause of these age-related cerebrovascular impairments remains incompletely understood. A characteristic feature of the aging vasculature is the increase in stiffness of the large elastic arteries. This increase in arterial stiffness is associated with elevated pulse pressure and blood flow pulsatility in the cerebral vasculature. Evidence from both humans and rodents supports that increases in large elastic artery stiffness are associated with cerebrovascular impairments. These impacts on cerebrovascular function are wide-ranging and include reductions in global and regional cerebral blood flow, cerebral small vessel disease, endothelial cell dysfunction, and impaired perivascular clearance. Furthermore, recent findings suggest that the relationship between arterial stiffness and cerebrovascular function may be influenced by genetics, specifically APOE and NOTCH genotypes. Given the strength of the evidence that age-related increases in arterial stiffness have deleterious impacts on the brain, interventions that target arterial stiffness are needed. The purpose of this review is to summarize the evidence from human and rodent studies, supporting the role of increased arterial stiffness in age-related cerebrovascular impairments.
Collapse
Affiliation(s)
- Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jill N Barnes
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - M Erin Moir
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
46
|
Dunaway LS, Luse MA, Nyshadham S, Bulut G, Alencar GF, Chavkin NW, Cortese-Krott M, Hirschi KK, Isakson BE. Obesogenic diet disrupts tissue-specific mitochondrial gene signatures in the artery and capillary endothelium. Physiol Genomics 2024; 56:113-127. [PMID: 37982169 PMCID: PMC11281809 DOI: 10.1152/physiolgenomics.00109.2023] [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: 09/21/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Endothelial cells (ECs) adapt to the unique needs of their resident tissue and metabolic perturbations, such as obesity. We sought to understand how obesity affects EC metabolic phenotypes, specifically mitochondrial gene expression. We investigated the mesenteric and adipose endothelium because these vascular beds have distinct roles in lipid homeostasis. Initially, we performed bulk RNA sequencing on ECs from mouse adipose and mesenteric vasculatures after a normal chow (NC) diet or high-fat diet (HFD) and found higher mitochondrial gene expression in adipose ECs compared with mesenteric ECs in both NC and HFD mice. Next, we performed single-cell RNA sequencing and categorized ECs as arterial, capillary, venous, or lymphatic. We found mitochondrial genes to be enriched in adipose compared with mesentery under NC conditions in artery and capillary ECs. After HFD, these genes were decreased in adipose ECs, becoming like mesenteric ECs. Transcription factor analysis revealed that peroxisome proliferator-activated receptor-γ (PPAR-γ) had high specificity in NC adipose artery and capillary ECs. These findings were recapitulated in single-nuclei RNA-sequencing data from human visceral adipose. The sum of these findings suggests that mesenteric and adipose arterial ECs metabolize lipids differently, and the transcriptional phenotype of the vascular beds converges in obesity due to downregulation of PPAR-γ in adipose artery and capillary ECs.NEW & NOTEWORTHY Using bulk and single-cell RNA sequencing on endothelial cells from adipose and mesentery, we found that an obesogenic diet induces a reduction in adipose endothelial oxidative phosphorylation gene expression, resulting in a phenotypic convergence of mesenteric and adipose endothelial cells. Furthermore, we found evidence that PPAR-γ drives this phenotypic shift. Mining of human data sets segregated based on body mass index supported these findings. These data point to novel mechanisms by which obesity induces endothelial dysfunction.
Collapse
Affiliation(s)
- Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Melissa A Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gamze Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gabriel F Alencar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Nicholas W Chavkin
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Miriam Cortese-Krott
- Department of Cardiology, Pneumology and Angiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Karen K Hirschi
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| |
Collapse
|
47
|
Eickelmann C, Lieder HR, Sturek M, Heusch G, Kleinbongard P. Differences in vasomotor function of mesenteric arteries between Ossabaw minipigs with predisposition to metabolic syndrome and Göttingen minipigs. Am J Physiol Heart Circ Physiol 2024; 326:H408-H417. [PMID: 38133620 PMCID: PMC11219054 DOI: 10.1152/ajpheart.00719.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/08/2023] [Accepted: 12/20/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome predisposes and contributes to the development and progression of atherosclerosis. The minipig strain "Ossabaw" is characterized by a predisposition to develop metabolic syndrome. We compared vasomotor function in Ossabaw minipigs before they developed their diseased phenotype to that of Göttingen minipigs without such genetic predisposition. Mesenteric arteries of adult Ossabaw and Göttingen minipigs were dissected postmortem and mounted on a myograph for isometric force measurements. Maximal vasoconstriction to potassium chloride (KClmax) was induced. Cumulative concentration-response curves were determined in response to norepinephrine. Endothelium-dependent (with carbachol) and endothelium-independent (with nitroprusside) vasodilation were analyzed after preconstriction by norepinephrine. In a bioinformatic analysis, variants/altered base pairs within genes associated with cardiovascular disease were analyzed. KClmax was similar between the minipig strains (15.6 ± 6.7 vs. 14.1 ± 3.4 ΔmN). Vasoconstriction in response to norepinephrine was more pronounced in Ossabaw than in Göttingen minipigs (increase of force to 143 ± 48 vs. 108 ± 38% of KClmax). Endothelium-dependent and endothelium-independent vasodilation were less pronounced in Ossabaw than in Göttingen minipigs (decrease of force to 46.4 ± 29.6 vs. 16.0 ± 18.4% and to 36.7 ± 25.2 vs. 2.3 ± 3.7% of norepinephrine-induced preconstriction). Vasomotor function was not different between the sexes. More altered base pairs/variants were identified in Ossabaw than in Göttingen minipigs for the exon encoding adrenoceptor-α1A. Vasomotor function in lean Ossabaw minipigs is shifted toward vasoconstriction and away from vasodilation in comparison with Göttingen minipigs, suggesting a genetic predisposition for vascular dysfunction and atherosclerosis in Ossabaw minipigs. Thus, Ossabaw minipigs may be a better model for human cardiovascular disease than Göttingen minipigs.NEW & NOTEWORTHY Animal models with a predisposition to metabolic syndrome and atherosclerosis are attracting growing interest for translational research, as they may better mimic the variability of patients with cardiovascular disease. In Ossabaw minipigs, with a polygenic predisposition to metabolic syndrome, but without the diseased phenotype, vasoconstriction is more and vasodilation is less pronounced in mesenteric arteries than in Göttingen minipigs. Ossabaw minipigs may be a more suitable model of human cardiovascular disease.
Collapse
Affiliation(s)
- Chantal Eickelmann
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Helmut Raphael Lieder
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Michael Sturek
- CorVus Biomedical, LLC, and CorVus Foundation, Inc., Crawfordsville, Indiana, United States
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| |
Collapse
|
48
|
Lucas-Herald AK, Montezano AC, Alves-Lopes R, Haddow L, O’Toole S, Flett M, Lee B, Amjad SB, Steven M, McNeilly J, Brooksbank K, Touyz RM, Ahmed SF. Effects of Sex Hormones on Vascular Reactivity in Boys With Hypospadias. J Clin Endocrinol Metab 2024; 109:e735-e744. [PMID: 37672642 PMCID: PMC10795938 DOI: 10.1210/clinem/dgad525] [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: 01/11/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Arteries from boys with hypospadias demonstrate hypercontractility and impaired vasorelaxation. The role of sex hormones in these responses in unclear. AIMS We compared effects of sex steroids on vascular reactivity in healthy boys and boys with hypospadias. METHODS Excess foreskin tissue was obtained from 11 boys undergoing hypospadias repair (cases) and 12 undergoing routine circumcision (controls) (median age [range], 1.5 [1.2-2.7] years) and small resistance arteries were isolated. Vessels were mounted on wire myographs and vascular reactivity was assessed in the absence/presence of 17β-estradiol, dihydrotestosterone (DHT), and testosterone. RESULTS In controls, testosterone and 17β-estradiol increased contraction (percent of maximum contraction [Emax]: 83.74 basal vs 125.4 after testosterone, P < .0002; and 83.74 vs 110.2 after estradiol, P = .02). 17β-estradiol reduced vasorelaxation in arteries from controls (Emax: 10.6 vs 15.6 to acetylcholine, P < .0001; and Emax: 14.6 vs 20.5 to sodium nitroprusside, P < .0001). In hypospadias, testosterone (Emax: 137.9 vs 107.2, P = .01) and 17β-estradiol (Emax: 156.9 vs 23.6, P < .0001) reduced contraction. Androgens, but not 17β-estradiol, increased endothelium-dependent and endothelium-independent vasorelaxation in cases (Emax: 77.3 vs 51.7 with testosterone, P = .02; and vs 48.2 with DHT to acetylcholine, P = .0001; Emax: 43.0 vs 39.5 with testosterone, P = .02; and 39.6 vs 37.5 with DHT to sodium nitroprusside, P = .04). CONCLUSION In healthy boys, testosterone and 17β-estradiol promote a vasoconstrictor phenotype, whereas in boys with hypospadias, these sex hormones reduce vasoconstriction, with androgens promoting vasorelaxation. Differences in baseline artery function may therefore be sex hormone-independent and the impact of early-life variations in androgen exposure on vascular function needs further study.
Collapse
Affiliation(s)
- Angela K Lucas-Herald
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, UK
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
- Research Institute of McGill University Health Center, McGill University, 1001 Boul Décarie, Montréal, QC H4A 3J1, Canada
| | - Rheure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Laura Haddow
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Stuart O’Toole
- Department of Pediatric Surgery, Royal Hospital for Children, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | - Martyn Flett
- Department of Pediatric Surgery, Royal Hospital for Children, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | - Boma Lee
- Department of Pediatric Surgery, Royal Hospital for Children, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | - S Basith Amjad
- Department of Pediatric Surgery, Royal Hospital for Children, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | - Mairi Steven
- Department of Pediatric Surgery, Royal Hospital for Children, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, Scotland, UK
| | - Jane McNeilly
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, UK
- Department of Clinical Biochemistry, Queen Elizabeth University Hospital, Glasgow G51 4TF, Scotland, UK
| | - Katriona Brooksbank
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Center for Research Excellence, University of Glasgow, 126 University Avenue, Glasgow G12 8TA, UK
- Research Institute of McGill University Health Center, McGill University, 1001 Boul Décarie, Montréal, QC H4A 3J1, Canada
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Royal Hospital for Children, 1345 Govan Road, Glasgow G51 4TF, UK
| |
Collapse
|
49
|
Moreau KL, Clayton ZS, DuBose LE, Rosenberry R, Seals DR. Effects of regular exercise on vascular function with aging: Does sex matter? Am J Physiol Heart Circ Physiol 2024; 326:H123-H137. [PMID: 37921669 PMCID: PMC11208002 DOI: 10.1152/ajpheart.00392.2023] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/11/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Vascular aging, featuring endothelial dysfunction and large elastic artery stiffening, is a major risk factor for the development of age-associated cardiovascular diseases (CVDs). Vascular aging is largely mediated by an excessive production of reactive oxygen species (ROS) and increased inflammation leading to reduced bioavailability of the vasodilatory molecule nitric oxide and remodeling of the arterial wall. Other cellular mechanisms (i.e., mitochondrial dysfunction, impaired stress response, deregulated nutrient sensing, cellular senescence), termed "hallmarks" or "pillars" of aging, may also contribute to vascular aging. Gonadal aging, which largely impacts women but also impacts some men, modulates the vascular aging process. Regular physical activity, including both aerobic and resistance exercise, is a first-line strategy for reducing CVD risk with aging. Although exercise is an effective intervention to counter vascular aging, there is considerable variation in the vascular response to exercise training with aging. Aerobic exercise improves large elastic artery stiffening in both middle-aged/older men and women and enhances endothelial function in middle-aged/older men by reducing oxidative stress and inflammation and preserving nitric oxide bioavailability; however, similar aerobic exercise training improvements are not consistently observed in estrogen-deficient postmenopausal women. Sex differences in adaptations to exercise may be related to gonadal aging and declines in estrogen in women that influence cellular-molecular mechanisms, disconnecting favorable signaling in the vasculature induced by exercise training. The present review will summarize the current state of knowledge on vascular adaptations to regular aerobic and resistance exercise with aging, the underlying mechanisms involved, and the moderating role of biological sex.
Collapse
Affiliation(s)
- Kerrie L Moreau
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Eastern Colorado Health Care System, Geriatric Research Education and Clinical Center, Aurora, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Lyndsey E DuBose
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Ryan Rosenberry
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
50
|
Shin J, Hong J, Edwards-Glenn J, Krukovets I, Tkachenko S, Adelus ML, Romanoski CE, Rajagopalan S, Podrez E, Byzova TV, Stenina-Adongravi O, Cherepanova OA. Unraveling the Role of Sex in Endothelial Cell Dysfunction: Evidence From Lineage Tracing Mice and Cultured Cells. Arterioscler Thromb Vasc Biol 2024; 44:238-253. [PMID: 38031841 PMCID: PMC10842863 DOI: 10.1161/atvbaha.123.319833] [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: 07/21/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Biological sex differences play a vital role in cardiovascular diseases, including atherosclerosis. The endothelium is a critical contributor to cardiovascular pathologies since endothelial cells (ECs) regulate vascular tone, redox balance, and inflammatory reactions. Although EC activation and dysfunction play an essential role in the early and late stages of atherosclerosis development, little is known about sex-dependent differences in EC. METHODS We used human and mouse aortic EC as well as EC-lineage tracing (Cdh5-CreERT2 Rosa-YFP [yellow fluorescence protein]) atherosclerotic Apoe-/- mice to investigate the biological sexual dimorphism of the EC functions in vitro and in vivo. Bioinformatics analyses were performed on male and female mouse aortic EC and human lung and aortic EC. RESULTS In vitro, female human and mouse aortic ECs showed more apoptosis and higher cellular reactive oxygen species levels than male EC. In addition, female mouse aortic EC had lower mitochondrial membrane potential (ΔΨm), lower TFAM (mitochondrial transcription factor A) levels, and decreased angiogenic potential (tube formation, cell viability, and proliferation) compared with male mouse aortic EC. In vivo, female mice had significantly higher lipid accumulation within the aortas, impaired glucose tolerance, and lower endothelial-mediated vasorelaxation than males. Using the EC-lineage tracing approach, we found that female lesions had significantly lower rates of intraplaque neovascularization and endothelial-to-mesenchymal transition within advanced atherosclerotic lesions but higher incidents of missing EC lumen coverage and higher levels of oxidative products and apoptosis. RNA-seq analyses revealed that both mouse and human female EC had higher expression of genes associated with inflammation and apoptosis and lower expression of genes related to angiogenesis and oxidative phosphorylation than male EC. CONCLUSIONS Our study delineates critical sex-specific differences in EC relevant to proinflammatory, pro-oxidant, and angiogenic characteristics, which are entirely consistent with a vulnerable phenotype in females. Our results provide a biological basis for sex-specific proatherosclerotic mechanisms.
Collapse
Affiliation(s)
- Junchul Shin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jonnelle Edwards-Glenn
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Irene Krukovets
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Maria L. Adelus
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
- Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, USA
| | - Casey E. Romanoski
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Sanjay Rajagopalan
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Eugene Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tatiana V. Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga Stenina-Adongravi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga A. Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|