101
|
Abstract
Kidney disease is associated with adverse consequences in many organs beyond the kidney, including the heart, lungs, brain, and intestines. The kidney-intestinal cross talk involves intestinal epithelial damage, dysbiosis, and generation of uremic toxins. Recent studies reveal that kidney injury expands the intestinal lymphatics, increases lymphatic flow, and alters the composition of mesenteric lymph. The intestinal lymphatics, like blood vessels, are a route for transporting potentially harmful substances generated by the intestines. The lymphatic architecture and actions are uniquely suited to take up and transport large macromolecules, functions that differentiate them from blood vessels, allowing them to play a distinct role in a variety of physiological and pathological processes. Here, we focus on the mechanisms by which kidney diseases result in deleterious changes in intestinal lymphatics and consider a novel paradigm of a vicious cycle of detrimental organ cross talk. This concept involves kidney injury-induced modulation of intestinal lymphatics that promotes production and distribution of harmful factors, which in turn contributes to disease progression in distant organ systems.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics (A.K.), Vanderbilt University Medical Center, Nashville, TN
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN (A.K.)
| | - Hai-Chun Yang
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Agnes B. Fogo
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine (A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Elaine L. Shelton
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Valentina Kon
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
102
|
Moulana M. Androgen-Induced Cardiovascular Risk in Polycystic Ovary Syndrome: The Role of T Lymphocytes. Life (Basel) 2023; 13:life13041010. [PMID: 37109539 PMCID: PMC10145997 DOI: 10.3390/life13041010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
An estimated 15-20% of reproductive-age women are affected by polycystic ovary syndrome (PCOS). PCOS is associated with substantial metabolic and cardiovascular long-term consequences. In young women with PCOS, several cardiovascular risk factors may be found, including chronic inflammation, high blood pressure, and elevated leukocytes. These women are at an increased risk of cardiovascular diseases (CVD), not only during the reproductive years, but also with aging and menopause; therefore, the early prevention and treatment of future cardiovascular adverse effects are necessary. The fundamental characteristic of PCOS is hyperandrogenemia, which is associated with increased pro-inflammatory cytokines and T lymphocytes. Whether these factors play a role in the pathophysiology of hypertension, a risk factor of CVD, due to PCOS is not well established. This review will briefly discuss how a modest increase in androgens in females is linked to the development of hypertension through pro-inflammatory cytokines and T lymphocyte subsets and the promotion of renal injury. Moreover, it reveals a few existing research gaps in this area, including the lack of specific therapy directed at androgen-induced inflammation and immune activation, thus emphasizing the necessity to explore the systemic inflammation in women with PCOS to halt the inevitable inflammatory process targeting the underlying abnormalities of CVD.
Collapse
Affiliation(s)
- Mohadetheh Moulana
- Department of Psychiatry and Human Behavior, Women's Health Research Center, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
103
|
Ahmad T, Ertuglu LA, Masenga SK, Kleyman TR, Kirabo A. The epithelial sodium channel in inflammation and blood pressure modulation. Front Cardiovasc Med 2023; 10:1130148. [PMID: 37123470 PMCID: PMC10132033 DOI: 10.3389/fcvm.2023.1130148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
A major regulator of blood pressure and volume homeostasis in the kidney is the epithelial sodium channel (ENaC). ENaC is composed of alpha(α)/beta(β)/gamma(γ) or delta(δ)/beta(β)/gamma(γ) subunits. The δ subunit is functional in the guinea pig, but not in routinely used experimental rodent models including rat or mouse, and thus remains the least understood of the four subunits. While the δ subunit is poorly expressed in the human kidney, we recently found that its gene variants are associated with blood pressure and kidney function. The δ subunit is expressed in the human vasculature where it may influence vascular function. Moreover, we recently found that the δ subunit is also expressed human antigen presenting cells (APCs). Our studies indicate that extracellular Na+ enters APCs via ENaC leading to inflammation and salt-induced hypertension. In this review, we highlight recent findings on the role of extra-renal ENaC in inflammation, vascular dysfunction, and blood pressure modulation. Targeting extra-renal ENaC may provide new drug therapies for salt-induced hypertension.
Collapse
Affiliation(s)
- Taseer Ahmad
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lale A. Ertuglu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sepiso K. Masenga
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
104
|
de la Visitación N, Chen W, Krishnan J, Van Beusecum JP, Amarnath V, Hennen EM, Zhao S, Saleem M, Ao M, Harrison DG, Patrick DM. Immunoproteasomal Processing of Isolevuglandin Adducts in Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536054. [PMID: 37383945 PMCID: PMC10299468 DOI: 10.1101/2023.04.10.536054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Isolevuglandins (isoLGs) are lipid aldehydes that form in the presence of reactive oxygen species (ROS) and drive immune activation. We found that isoLG-adducts are presented within the context of major histocompatibility complexes (MHC-I) by an immunoproteasome dependent mechanism. Pharmacologic inhibition of LMP7, the chymotrypsin subunit of the immunoproteasome, attenuates hypertension and tissue inflammation in the angiotensin II (Ang II) model of hypertension. Genetic loss of function of all immunoproteasome subunits or conditional deletion of LMP7 in dendritic cell (DCs) or endothelial cells (ECs) attenuated hypertension, reduced aortic T cell infiltration, and reduced isoLG-adduct MHC-I interaction. Furthermore, isoLG adducts structurally resemble double-stranded DNA and contribute to the activation of STING in ECs. These studies define a critical role of the immunoproteasome in the processing and presentation of isoLG-adducts. Moreover they define a role of LMP7 as a regulator of T cell activation and tissue infiltration in hypertension.
Collapse
|
105
|
Fehrenbach DJ, Nguyen B, Alexander MR, Madhur MS. Modulating T Cell Phenotype and Function to Treat Hypertension. KIDNEY360 2023; 4:e534-e543. [PMID: 36951464 PMCID: PMC10278787 DOI: 10.34067/kid.0000000000000090] [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: 02/11/2022] [Accepted: 01/25/2023] [Indexed: 03/24/2023]
Abstract
Hypertension is the leading modifiable risk factor of worldwide morbidity and mortality because of its effects on cardiovascular and renal end-organ damage. Unfortunately, BP control is not sufficient to fully reduce the risks of hypertension, underscoring the need for novel therapies that address end-organ damage in hypertension. Over the past several decades, the link between immune activation and hypertension has been well established, but there are still no therapies for hypertension that specifically target the immune system. In this review, we describe the critical role played by T cells in hypertension and hypertensive end-organ damage and outline potential therapeutic targets to modulate T-cell phenotype and function in hypertension without causing global immunosuppression.
Collapse
Affiliation(s)
- Daniel J. Fehrenbach
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Bianca Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| |
Collapse
|
106
|
Benson LN, Guo Y, Deck K, Mora C, Liu Y, Mu S. The link between immunity and hypertension in the kidney and heart. Front Cardiovasc Med 2023; 10:1129384. [PMID: 36970367 PMCID: PMC10034415 DOI: 10.3389/fcvm.2023.1129384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Hypertension is the primary cause of cardiovascular disease, which is a leading killer worldwide. Despite the prevalence of this non-communicable disease, still between 90% and 95% of cases are of unknown or multivariate cause ("essential hypertension"). Current therapeutic options focus primarily on lowering blood pressure through decreasing peripheral resistance or reducing fluid volume, but fewer than half of hypertensive patients can reach blood pressure control. Hence, identifying unknown mechanisms causing essential hypertension and designing new treatment accordingly are critically needed for improving public health. In recent years, the immune system has been increasingly implicated in contributing to a plethora of cardiovascular diseases. Many studies have demonstrated the critical role of the immune system in the pathogenesis of hypertension, particularly through pro-inflammatory mechanisms within the kidney and heart, which, eventually, drive a myriad of renal and cardiovascular diseases. However, the precise mechanisms and potential therapeutic targets remain largely unknown. Therefore, identifying which immune players are contributing to local inflammation and characterizing pro-inflammatory molecules and mechanisms involved will provide promising new therapeutic targets that could lower blood pressure and prevent progression from hypertension into renal or cardiac dysfunction.
Collapse
Affiliation(s)
- Lance N. Benson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| | | | | | | | | | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, United States
| |
Collapse
|
107
|
Van Beusecum JP, Rianto F, Teakell J, Kon V, Sparks MA, Hoorn EJ, Kirabo A, Ramkumar N. Novel Concepts in Nephron Sodium Transport: A Physiological and Clinical Perspective. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:124-136. [PMID: 36868728 DOI: 10.1053/j.akdh.2022.12.007] [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: 09/20/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 04/13/2023]
Abstract
The kidneys play a critical role in maintaining total body sodium (Na+) balance across a wide range of dietary intake, accomplished by a concerted effort involving multiple Na+ transporters along the nephron. Furthermore, nephron Na+ reabsorption and urinary Na+ excretion are closely linked to renal blood flow and glomerular filtration such that perturbations in either of them can modify Na+ transport along the nephron, ultimately resulting in hypertension and other Na+-retentive states. In this article, we provide a brief physiological overview of nephron Na+ transport and illustrate clinical syndromes and therapeutic agents that affect Na+ transporter function. We highlight recent advances in kidney Na+ transport, particularly the role of immune cells, lymphatics, and interstitial Na+ in regulating Na+ reabsorption, the emergence of potassium (K+) as a regulator of Na+ transport, and the evolution of the nephron to modulate Na+ transport.
Collapse
Affiliation(s)
- Justin P Van Beusecum
- Ralph H. Johnson VA Medical Center, Charleston, SC; Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Fitra Rianto
- Division of Nephrology, Department of Medicine, Duke University School of Medicine and Renal Section, Durham VA Health Care System Durham, Durham, NC
| | - Jade Teakell
- Division of Renal Diseases and Hypertension, Department of Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, TX
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine and Renal Section, Durham VA Health Care System Durham, Durham, NC
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Nirupama Ramkumar
- Division of Nephrology and Hypertension, Department of Medicine, University of Utah Health, Salt Lake City, UT.
| |
Collapse
|
108
|
Alexander MR, Dale BL, Smart CD, Elijovich F, Wogsland CE, Lima SM, Irish JM, Madhur MS. Immune Profiling Reveals Decreases in Circulating Regulatory and Exhausted T Cells in Human Hypertension. JACC Basic Transl Sci 2023; 8:319-336. [PMID: 37034287 PMCID: PMC10077123 DOI: 10.1016/j.jacbts.2022.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 01/06/2023]
Abstract
Evidence from nonhuman animal models demonstrates an important role for immune cells in hypertension, but immune cell changes in human hypertension are less clear. Using mass cytometry, we demonstrate novel and selective reductions in CCR10+ regulatory T cells (Tregs) and PD-1+CD57-CD8+ memory T cells. RNA sequencing reveals that CCR10+ Tregs exhibit gene expression changes consistent with enhanced immunosuppressive function. In addition, CITE-Seq demonstrates that PD-1+CD57-CD8+ memory T cells exhibit features of T-cell exhaustion. Taken together, these results provide novel evidence for decreases in anti-inflammatory and/or hypofunctional T-cell populations that may contribute to enhanced inflammation in human hypertension.
Collapse
Affiliation(s)
- Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Bethany L. Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Current affiliation: Pirche, Berlin, Germany
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cara E. Wogsland
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Current affiliation: KinN Therapeutics, Bergen, Norway
| | - Sierra M. Lima
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan M. Irish
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
109
|
Ekperikpe US, Poudel B, Shields CA, Mandal S, Cornelius DC, Williams JM. Neutralizing MIP3 α Reduces Renal Immune Cell Infiltration and Progressive Renal Injury in Young Obese Dahl Salt-Sensitive Rats. J Pharmacol Exp Ther 2023; 384:445-454. [PMID: 36507846 PMCID: PMC9976792 DOI: 10.1124/jpet.122.001298] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Recently, we reported that the early progression of renal injury in obese Dahl salt-sensitive leptin receptor mutant (SSLepRmutant) rats was associated with increased macrophage inflammatory protein 3-α (MIP3α) expression prior to puberty. Therefore, this study tested the hypothesis that MIP3α plays a role in recruiting immune cells, thereby triggering renal inflammation and early progressive renal injury in SSLepRmutant rats prior to puberty. Four-week-old Dahl salt-sensitive (SS) and SSLepRmutant rats either served as control (IgG; intraperitoneal, every other day) or received MIP3α-neutralizing antibody (MNA; 100 µg/kg) for 4 weeks. MNA reduced circulating and renal MIP3α levels and proinflammatory immune cells by 50%. Although MNA treatment did not affect blood glucose and plasma cholesterol levels, MNA markedly decreased insulin resistance and triglyceride levels in SSLepRmutant rats. We observed no differences in mean arterial pressure (MAP) between SS and SSLepRmutant rats, and MNA had no effect on MAP in either strain. Proteinuria was significantly increased in SSLepRmutant rats versus SS rats over the course of the study. Treatment with MNA markedly decreased proteinuria in SSLepRmutant rats while not affecting SS rats. Also, MNA decreased glomerular and tubular injury and renal fibrosis in SSLepRmutant rats while not affecting SS rats. Overall, these data indicate that MIP3α plays an important role in renal inflammation during the early progression of renal injury in obese SSLepRmutant rats prior to puberty. These data also suggest that MIP3α may be a novel therapeutic target to inhibit insulin resistance and prevent progressive proteinuria in obese children. SIGNIFICANCE STATEMENT: Childhood obesity is increasing at an alarming rate and is now being associated with renal disease. Although most studies have focused on the mechanisms of renal injury associated with adult obesity, few studies have examined the mechanisms of renal injury involved during childhood obesity. In the current study, we observed that the progression of renal injury in obese Dahl salt-sensitive leptin receptor mutant rats was associated with an increase in MIP3α, a chemokine, before puberty, and inhibition of MIP3α markedly reduced renal injury.
Collapse
Affiliation(s)
- Ubong S Ekperikpe
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bibek Poudel
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sautan Mandal
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Departments of Pharmacology and Toxicology and Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
110
|
Abstract
A large body of evidence has emerged in the past decade supporting a role for the gut microbiome in the regulation of blood pressure. The field has moved from association to causation in the last 5 years, with studies that have used germ-free animals, antibiotic treatments and direct supplementation with microbial metabolites. The gut microbiome can regulate blood pressure through several mechanisms, including through gut dysbiosis-induced changes in microbiome-associated gene pathways in the host. Microbiota-derived metabolites are either beneficial (for example, short-chain fatty acids and indole-3-lactic acid) or detrimental (for example, trimethylamine N-oxide), and can activate several downstream signalling pathways via G protein-coupled receptors or through direct immune cell activation. Moreover, dysbiosis-associated breakdown of the gut epithelial barrier can elicit systemic inflammation and disrupt intestinal mechanotransduction. These alterations activate mechanisms that are traditionally associated with blood pressure regulation, such as the renin-angiotensin-aldosterone system, the autonomic nervous system, and the immune system. Several methodological and technological challenges remain in gut microbiome research, and the solutions involve minimizing confounding factors, establishing causality and acting globally to improve sample diversity. New clinical trials, precision microbiome medicine and computational methods such as Mendelian randomization have the potential to enable leveraging of the microbiome for translational applications to lower blood pressure.
Collapse
|
111
|
Life is really simple, but we insist on making it complicated. (Confucius). Hypertens Res 2023; 46:1051-1053. [PMID: 36759664 DOI: 10.1038/s41440-023-01212-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/11/2023]
|
112
|
Rodriguez-Iturbe B, Johnson RJ, Sanchez-Lozada LG, Pons H. HSP70 and Primary Arterial Hypertension. Biomolecules 2023; 13:272. [PMID: 36830641 PMCID: PMC9953434 DOI: 10.3390/biom13020272] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Heat shock protein 70 (HSP70) production is a stress-generated cellular response with high interspecies homology. HSP70 has both chaperone and cytokine functions and may induce, depending on the context, tolerogenic anti-inflammatory reactivity or immunogenic and autoimmune reactivity. Intracellular (chaperoning transit of antigens to MHC in antigen-presenting cells) and extracellular HSP70-related effects are associated with hypertension, which is an inflammatory condition recognized as the most important risk factor for cardiovascular disease mortality. Here, we review (a) the relationship between HSP70, inflammation and immune reactivity, (b) clinical evidence relating to stress, HSP70 and anti-HSP70 reactivity with primary hypertension and (c) experimental data showing that salt-sensitive hypertension is associated with delayed hypersensitivity to HSP70. This is a consequence of anti-HSP70 reactivity in the kidneys and may be prevented and corrected by the T-cell-driven inhibition of kidney inflammation triggered by specific epitopes of HSP70. Finally, we discuss our postulate that lifelong stress signals and danger-associated molecular patterns stimulate HSP-70 and individual genetic and epigenetic characteristics determine whether the HSP70 response would drive inflammatory immune reactivity causing hypertension or, alternatively, would drive immunomodulatory responses that protect against hypertension.
Collapse
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laura Gabriela Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico
| | - Hector Pons
- Facultad de Medicina, Universidad del Zulia, Maracaibo 4011, Venezuela
| |
Collapse
|
113
|
Abstract
Several clinical and large population studies indicate that women are more salt-sensitive than men, yet the precise mechanisms by which the sexually dimorphic onset manifests remains incompletely understood. Here, we evaluate recent epidemiological data and highlight current knowledge from studies investigating sex-specific mechanisms of salt-sensitive blood pressure (SSBP). Emerging evidence indicates that women of all ethnicities are more salt-sensitive than men, at all ages both premenopausal and postmenopausal. However, menopause exacerbates severity and prevalence of SSBP, suggesting that female sex chromosomes predispose to and female sex hormones mitigate SSBP. Results from both human and rodent studies support the contribution of enhanced and inappropriate activation of the aldosterone-ECMR (endothelial cell mineralocorticoid receptor) axis promoting vascular dysfunction in females. Increases in adrenal response to angiotensin II, in association with higher ECMR expression and activation of endothelial ENaC (epithelial sodium channel) in females compared to males, are emerging as central players in the development of endothelial dysfunction and SSBP in females. Female sex increases the prevalence and susceptibility of SSBP and sex hormones and sex chromosome complement may exert antagonistic effects in the development of the female heightened SSBP.
Collapse
Affiliation(s)
- Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jessica L. Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Physiology Department, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J. Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
114
|
Martin K, Toussaint ND, Tan SJ, Hewitson TD. Skin regulation of salt and blood pressure and potential clinical implications. Hypertens Res 2023; 46:408-416. [PMID: 36434290 DOI: 10.1038/s41440-022-01096-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Sodium chloride, as salt, gives rise to hypertension. Nevertheless, individual susceptibility to the ramifications of sodium chloride is heterogeneous. The conventional nephron-centric regulation of sodium with neurohormonal inputs and responses is now expanded to include an intricate extrarenal pathway including the endothelium, skin, lymphatics, and immune cells. An overabundance of sodium is buffered and regulated by the skin interstitium. Excess sodium passes through (and damages) the vascular endothelium and can be dynamically stored in the skin, modulated by skin immune cells and lymphatics. This excess interstitially stored sodium is implicated in hypertension, cardiovascular dysfunction, metabolic disruption, and inflammatory dysregulation. This extrarenal pathway of regulating sodium represents a novel target for better blood pressure management, rebalancing disturbed inflammation, and hence addressing cardiovascular and metabolic disease.
Collapse
Affiliation(s)
- Kylie Martin
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia. .,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.
| | - Nigel D Toussaint
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Sven-Jean Tan
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
115
|
Gobert AP, Asim M, Smith TM, Williams KJ, Barry DP, Allaman MM, McNamara KM, Hawkins CV, Delgado AG, Piazuelo MB, Rathmacher JA, Wilson KT. The nutraceutical electrophile scavenger 2-hydroxybenzylamine (2-HOBA) attenuates gastric cancer development caused by Helicobacter pylori. Biomed Pharmacother 2023; 158:114092. [PMID: 36493697 PMCID: PMC9879697 DOI: 10.1016/j.biopha.2022.114092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/22/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Stomach cancer is a leading cause of cancer death. Helicobacter pylori is a bacterial gastric pathogen that is the primary risk factor for carcinogenesis, associated with its induction of inflammation and DNA damage. Dicarbonyl electrophiles are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. 2-hydroxybenzylamine (2-HOBA) is a natural compound derived from buckwheat seeds and acts as a potent scavenger of reactive aldehydes. Our goal was to investigate the effect of 2-HOBA on the pathogenesis of H. pylori infection. We used transgenic FVB/N insulin-gastrin (INS-GAS) mice as a model of gastric cancer. First, we found that 2-HOBA is bioavailable in the gastric tissues of these mice after supplementation in the drinking water. Moreover, 2-HOBA reduced the development of gastritis in H. pylori-infected INS-GAS mice without affecting the bacterial colonization level in the stomach. Further, we show that the development of gastric dysplasia and carcinoma was significantly reduced by 2-HOBA. Concomitantly, DNA damage were also inhibited by 2-HOBA treatment in H. pylori-infected mice. In parallel, DNA damage was inhibited by 2-HOBA in H. pylori-infected gastric epithelial cells in vitro. In conclusion, 2-HOBA, which has been shown to be safe in human clinical trials, represents a promising nutritional compound for the chemoprevention of the more severe effects of H. pylori infection.
Collapse
Affiliation(s)
- Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thaddeus M. Smith
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kamery J. Williams
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret M. Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kara M. McNamara
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V. Hawkins
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John A. Rathmacher
- MTI BioTech, Iowa State University Research Park, Ames, IA, USA,Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA,Correspondence to: Vanderbilt University Medical Center, 2215B Garland Ave., 1030C MRB IV, Nashville, TN 37232-0252, USA. (K.T. Wilson)
| |
Collapse
|
116
|
Caspa Gokulan R, Paulrasu K, Azfar J, El-Rifai W, Que J, Boutaud OG, Ban Y, Gao Z, Buitrago MG, Dikalov SI, Zaika AI. Protein adduction causes non-mutational inhibition of p53 tumor suppressor. Cell Rep 2023; 42:112024. [PMID: 36848235 PMCID: PMC9989503 DOI: 10.1016/j.celrep.2023.112024] [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: 04/05/2022] [Revised: 06/04/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
p53 is a key tumor suppressor that is frequently mutated in human tumors. In this study, we investigated how p53 is regulated in precancerous lesions prior to mutations in the p53 gene. Analyzing esophageal cells in conditions of genotoxic stress that promotes development of esophageal adenocarcinoma, we find that p53 protein is adducted with reactive isolevuglandins (isoLGs), products of lipid peroxidation. Modification of p53 protein with isoLGs diminishes its acetylation and binding to the promoters of p53 target genes causing modulation of p53-dependent transcription. It also leads to accumulation of adducted p53 protein in intracellular amyloid-like aggregates that can be inhibited by isoLG scavenger 2-HOBA in vitro and in vivo. Taken together, our studies reveal a posttranslational modification of p53 protein that causes molecular aggregation of p53 protein and its non-mutational inactivation in conditions of DNA damage that may play an important role in human tumorigenesis.
Collapse
Affiliation(s)
| | | | - Jamal Azfar
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Olivier G Boutaud
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuguang Ban
- Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | - Zhen Gao
- Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | | | - Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander I Zaika
- Department of Surgery, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA.
| |
Collapse
|
117
|
The Effect of Renal Denervation on T Cells in Patients with Resistant Hypertension. Int J Mol Sci 2023; 24:ijms24032493. [PMID: 36768814 PMCID: PMC9917284 DOI: 10.3390/ijms24032493] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
(1) Background: Sympathetic overactivity is a major contributor to resistant hypertension (RH). According to animal studies, sympathetic overactivity increases immune responses, thereby aggravating hypertension and cardiovascular outcomes. Renal denervation (RDN) reduces sympathetic nerve activity in RH. Here, we investigate the effect of RDN on T-cell signatures in RH. (2) Methods: Systemic inflammation and T-cell subsets were analyzed in 17 healthy individuals and 30 patients with RH at baseline and 6 months after RDN. (3) Results: The patients with RH demonstrated higher levels of pro-inflammatory cytokines and higher frequencies of CD4+ effector memory (TEM), CD4+ effector memory residential (TEMRA) and CD8+ central memory (TCM) cells than the controls. After RDN, systolic automated office blood pressure (BP) decreased by -17.6 ± 18.9 mmHg. Greater BP reductions were associated with higher CD4+ TEM (r -0.421, p = 0.02) and CD8+ TCM (r -0.424, p = 0.02) frequencies at baseline. The RDN responders, that is, the patients with ≥10mmHg systolic BP reduction, showed reduced pro-inflammatory cytokine levels, whereas the non-responders had unchanged inflammatory activity and higher CD8+ TEMRA frequencies with increased cellular cytokine production. (4) Conclusions: The pro-inflammatory state of patients with RH is characterized by altered T-cell signatures, especially in non-responders. A detailed analysis of T cells might be useful in selecting patients for RDN.
Collapse
|
118
|
Mass Cytometry Reveals the Imbalanced Immune State in the Peripheral Blood of Patients with Essential Hypertension. Cardiovasc Ther 2023; 2023:9915178. [PMID: 36891527 PMCID: PMC9988372 DOI: 10.1155/2023/9915178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 03/02/2023] Open
Abstract
Mounting evidence has confirmed that essential hypertension (EH) is closely related to low-grade inflammation, but there is still a lack of in-depth understanding of the state of immune cells in the circulating blood of patients with EH. We analyzed whether hypertensive peripheral blood immune cell balance was destroyed. The peripheral blood mononuclear cells (PBMCs) of all subjects were analyzed using time-of-flight cytometry (CyTOF) based on 42 kinds of metal-binding antibodies. CD45+ cells were categorized into 32 kinds of subsets. Compared with the health control (HC) group, the percentage of total dendritic cells, two kinds of myeloid dendritic cell subsets, one intermediate/nonclassical monocyte subset and one CD4+ central memory T cell subset in the EH group, was significantly higher; the percentage of low-density neutrophils, four kinds of classical monocyte subsets, one CD14lowCD16- monocyte subset, one naive CD4+ and one naive CD8+ T cell subsets, one CD4+ effector and one CD4+ central memory T cell subsets, one CD8+ effector memory T cell subset, and one terminally differentiated γδ T cell subset, decreased significantly in EH. What is more, the expression of many important antigens was enhanced in CD45+ immune cells, granulocytes, and B cells in patients with EH. In conclusion, the altered number and antigen expression of immune cells reflect the imbalanced immune state of the peripheral blood in patients with EH.
Collapse
|
119
|
Ertuglu LA, Laffer CL, Kirabo A. In Memoriam: Fernando Elijovich. Hypertension 2023; 80:e1-e3. [PMID: 36475860 DOI: 10.1161/hypertensionaha.122.20541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lale A Ertuglu
- Division of Nephrology, Department of Medicine (L.A.E.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
120
|
Wienecke LM, Leid JM, Leuschner F, Lavine KJ. Imaging Targets to Visualize the Cardiac Immune Landscape in Heart Failure. Circ Cardiovasc Imaging 2023; 16:e014071. [PMID: 36649453 PMCID: PMC9858350 DOI: 10.1161/circimaging.122.014071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Heart failure involves a complex interplay between diverse populations of immune cells that dynamically shift across the natural history of disease. Within this context, the character of the immune response is a key determinant of clinical outcomes. Recent technological advances in single-cell transcriptomic, spatial, and proteomic technologies have fueled an explosion of new and clinically relevant insights into distinct immune cell populations that reside within the diseased heart including potential targets for molecular imaging and therapy. In this review, we will discuss the immune cell types and their respective functions with respect to myocardial infarction remodeling, dilated cardiomyopathy, and heart failure with preserved ejection fraction. In addition, we give a brief overview regarding myocarditis and cardiac sarcoidosis as inflammatory heart failure etiologies. We will highlight markers and cell populations as targets for molecular imaging to visualize inflammation and tissue healing and discuss clinical implications including the development and implementation of precision medicine approaches.
Collapse
Affiliation(s)
- Laura M. Wienecke
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Jamison M. Leid
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Florian Leuschner
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Center for Regenerative Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
121
|
Benson LN, Liu Y, Deck K, Mora C, Mu S. IFN- γ Contributes to the Immune Mechanisms of Hypertension. KIDNEY360 2022; 3:2164-2173. [PMID: 36591357 PMCID: PMC9802558 DOI: 10.34067/kid.0001292022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/19/2022] [Indexed: 12/31/2022]
Abstract
Hypertension is the leading cause of cardiovascular disease and the primary risk factor for mortality worldwide. For more than half a century, researchers have demonstrated that immunity plays an important role in the development of hypertension; however, the precise mechanisms are still under investigation. The current body of knowledge indicates that proinflammatory cytokines may play an important role in contributing to immune-related pathogenesis of hypertension. Interferon gamma (IFN-γ), in particular, as an important cytokine that modulates immune responses, has been recently identified as a critical regulator of blood pressure by several groups, including us. In this review, we focus on exploring the role of IFN-γ in contributing to the pathogenesis of hypertension, outlining the various immune producers of this cytokine and described signaling mechanisms involved. We demonstrate a key role for IFN-γ in hypertension through global knockout studies and related downstream signaling pathways that IFN-γ production from CD8+ T cell (CD8T) in the kidney promoting CD8T-stimulated salt retention via renal tubule cells, thereby exacerbating hypertension. We discuss potential activators of these T cells described by the current literature and relay a novel hypothesis for activation.
Collapse
Affiliation(s)
- Lance N. Benson
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Yunmeng Liu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Katherine Deck
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Christoph Mora
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
122
|
Ertuglu LA, Mutchler AP, Yu J, Kirabo A. Inflammation and oxidative stress in salt sensitive hypertension; The role of the NLRP3 inflammasome. Front Physiol 2022; 13:1096296. [PMID: 36620210 PMCID: PMC9814168 DOI: 10.3389/fphys.2022.1096296] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Salt-sensitivity of blood pressure is an independent risk factor for cardiovascular disease and affects approximately half of the hypertensive population. While the precise mechanisms of salt-sensitivity remain unclear, recent findings on body sodium homeostasis and salt-induced immune cell activation provide new insights into the relationship between high salt intake, inflammation, and hypertension. The immune system, specifically antigen-presenting cells (APCs) and T cells, are directly implicated in salt-induced renal and vascular injury and hypertension. Emerging evidence suggests that oxidative stress and activation of the NLRP3 inflammasome drive high sodium-mediated activation of APCs and T cells and contribute to the development of renal and vascular inflammation and hypertension. In this review, we summarize the recent insights into our understanding of the mechanisms of salt-sensitive hypertension and discuss the role of inflammasome activation as a potential therapeutic target.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United Staes,*Correspondence: Annet Kirabo, ; Lale A. Ertuglu,
| | - Ashley Pitzer Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Justin Yu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States,*Correspondence: Annet Kirabo, ; Lale A. Ertuglu,
| |
Collapse
|
123
|
Bloodworth N, Barbaro NR, Moretti R, Harrison DG, Meiler J. Rosetta FlexPepDock to predict peptide-MHC binding: An approach for non-canonical amino acids. PLoS One 2022; 17:e0275759. [PMID: 36512534 PMCID: PMC9746977 DOI: 10.1371/journal.pone.0275759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/22/2022] [Indexed: 12/15/2022] Open
Abstract
Computation methods that predict the binding of peptides to MHC-I are important tools for screening and identifying immunogenic antigens and have the potential to accelerate vaccine and drug development. However, most available tools are sequence-based and optimized only for peptides containing the twenty canonical amino acids. This omits a large number of peptides containing non-canonical amino acids (NCAA), or residues that undergo varied post-translational modifications such as glycosylation or phosphorylation. These modifications fundamentally alter peptide immunogenicity. Similarly, existing structure-based methods are biased towards canonical peptide backbone structures, which may or may not be preserved when NCAAs are present. Rosetta FlexPepDock ab-initio is a structure-based computational protocol able to evaluate peptide-receptor interaction where no prior information of the peptide backbone is known. We benchmarked FlexPepDock ab-initio for docking canonical peptides to MHC-I, and illustrate for the first time the method's ability to accurately model MHC-I bound epitopes containing NCAAs. FlexPepDock ab-initio protocol was able to recapitulate near-native structures (≤1.5Å) in the top lowest-energy models for 20 out of 25 cases in our initial benchmark. Using known experimental binding affinities of twenty peptides derived from an influenza-derived peptide, we showed that FlexPepDock protocol is able to predict relative binding affinity as Rosetta energies correlate well with experimental values (r = 0.59, p = 0.006). ROC analysis revealed 80% true positive and a 40% false positive rate, with a prediction power of 93%. Finally, we demonstrate the protocol's ability to accurately recapitulate HLA-A*02:01 bound phosphopeptide backbone structures and relative binding affinity changes, the theoretical structure of the lymphocytic choriomeningitis derived glycosylated peptide GP392 bound to MHC-I H-2Db, and isolevuglandin-adducted peptides. The ability to use non-canonical amino acids in the Rosetta FlexPepDock protocol may provide useful insight into critical amino acid positions where the post-translational modification modulates immunologic responses.
Collapse
Affiliation(s)
- Nathaniel Bloodworth
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Natália Ruggeri Barbaro
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Rocco Moretti
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| |
Collapse
|
124
|
Goodlett BL, Balasubbramanian D, Navaneethabalakrishnan S, Love SE, Luera EM, Konatham S, Chiasson VL, Wedgeworth S, Rutkowski JM, Mitchell BM. Genetically inducing renal lymphangiogenesis attenuates hypertension in mice. Clin Sci (Lond) 2022; 136:1759-1772. [PMID: 36345993 PMCID: PMC10586591 DOI: 10.1042/cs20220547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Hypertension (HTN) is associated with renal proinflammatory immune cell infiltration and increased sodium retention. We reported previously that renal lymphatic vessels, which are responsible for trafficking immune cells from the interstitial space to draining lymph nodes, increase in density under hypertensive conditions. We also demonstrated that augmenting renal lymphatic density can prevent HTN in mice. Whether renal lymphangiogenesis can treat HTN in mice is unknown. We hypothesized that genetically inducing renal lymphangiogenesis after the establishment of HTN would attenuate HTN in male and female mice from three different HTN models. METHODS Mice with inducible kidney-specific overexpression of VEGF-D (KidVD) experience renal lymphangiogenesis upon doxycycline administration. HTN was induced in KidVD+ and KidVD- mice by subcutaneous release of angiotensin II, administration of the nitric oxide synthase inhibitor L-NAME, or consumption of a 4% salt diet following a L-NAME priming and washout period. After a week of HTN stimuli treatment, doxycycline was introduced. Systolic blood pressure (SBP) readings were taken weekly. Kidney function was determined from urine and serum measures. Kidneys were processed for RT-qPCR, flow cytometry, and imaging. RESULTS Mice that underwent renal-specific lymphangiogenesis had significantly decreased SBP and renal proinflammatory immune cells. Additionally, renal lymphangiogenesis was associated with a decrease in sodium transporter expression and increased fractional excretion of sodium, indicating improved sodium handling efficiency. CONCLUSIONS These findings demonstrate that augmenting renal lymphangiogenesis can treat HTN in male and female mice by improving renal immune cell trafficking and sodium handling.
Collapse
Affiliation(s)
- Bethany L Goodlett
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | | | | | - Sydney E Love
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Emily M Luera
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Sunitha Konatham
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Valorie L Chiasson
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Sophie Wedgeworth
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Joseph M Rutkowski
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| |
Collapse
|
125
|
Influenza Virus Infection during Pregnancy as a Trigger of Acute and Chronic Complications. Viruses 2022; 14:v14122729. [PMID: 36560733 PMCID: PMC9786233 DOI: 10.3390/v14122729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) infection during pregnancy disrupts maternal and fetal health through biological mechanisms, which are to date poorly characterised. During pregnancy, the viral clearance mechanisms from the lung are sub-optimal and involve hyperactive innate and adaptive immune responses that generate wide-spread inflammation. Pregnancy-related adaptations of the immune and the cardiovascular systems appear to result in delayed recovery post-viral infection, which in turn promotes a prolonged inflammatory phenotype, increasing disease severity, and causing maternal and fetal health problems. This has immediate and long-term consequences for the mother and fetus, with complications including acute cardiopulmonary distress syndrome in the mother that lead to perinatal complications such as intrauterine growth restriction (IUGR), and birth defects; cleft lip, cleft palate, neural tube defects and congenital heart defects. In addition, an increased risk of long-term neurological disorders including schizophrenia in the offspring is reported. In this review we discuss the pathophysiology of IAV infection during pregnancy and its striking similarity to other well-established complications of pregnancy such as preeclampsia. We discuss general features of vascular disease with a focus on vascular inflammation and define the "Vascular Storm" that is triggered by influenza infection during pregnancy, as a pivotal disease mechanism for short and long term cardiovascular complications.
Collapse
|
126
|
Zhao M, Zheng Z, Li C, Wan J, Wang M. Developmental endothelial locus-1 in cardiovascular and metabolic diseases: A promising biomarker and therapeutic target. Front Immunol 2022; 13:1053175. [PMID: 36518760 PMCID: PMC9742254 DOI: 10.3389/fimmu.2022.1053175] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular and metabolic diseases (CVMDs) are a leading cause of death worldwide and impose a major socioeconomic burden on individuals and healthcare systems, underscoring the urgent need to develop new drug therapies. Developmental endothelial locus-1 (DEL-1) is a secreted multifunctional domain protein that can bind to integrins and play an important role in the occurrence and development of various diseases. Recently, DEL-1 has attracted increased interest for its pharmacological role in the treatment and/or management of CVMDs. In this review, we present the current knowledge on the predictive and therapeutic role of DEL-1 in a variety of CVMDs, such as atherosclerosis, hypertension, cardiac remodeling, ischemic heart disease, obesity, and insulin resistance. Collectively, DEL-1 is a promising biomarker and therapeutic target for CVMDs.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China,Cardiovascular Research Institute, Wuhan University, Wuhan, China,Hubei Key Laboratory of Cardiology, Wuhan, China,*Correspondence: Menglong Wang, ; Jun Wan,
| |
Collapse
|
127
|
Harimenshi D, Niyongabo T, Preux PM, Aboyans V, Desormais I. Hypertension and associated factors in HIV-infected patients receiving antiretroviral treatment in Burundi: a cross-sectional study. Sci Rep 2022; 12:20509. [PMID: 36443478 PMCID: PMC9705296 DOI: 10.1038/s41598-022-24997-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Currently, the life expectancy of people living with the human immunodeficiency virus (HIV) and the general population are similar. Hypertension is a major public health issue in Africa and is largely underdiagnosed. Most HIV-infected individuals, especially those on Anti-Retroviral Therapy (ART) have hypertension. Our project aims to determine the prevalence of hypertension and associated factors amongst HIV-infected adults treated by ART in Burundi. A cross-sectional study was conducted among HIV-infected subjects over the age of 20, managed in five healthcare centers for people living with HIV (PLWH). The World Health Organization STEPWISE survey and anthropometric measurements were employed. Blood pressure was measured according to the ESC 2018 recommendations. 1 250 HIV-infected patients aged between 35.4 and 50.2 years were included (18.4% men). The prevalence of hypertension was 17.4% (95% CI 13.2-22.1). Approximately 47.25% of HIV patients with hypertension were previously undiagnosed. Other factors were associated with HTN, such as being overweight (OR 2.88; 95% CI 1.46-5.62), obesity (OR 2.65; 95% CI 1.27-5.55), longer duration of HIV infection: ≥ 10 years (OR 1.04; 95% CI 1.14-3.20), diabetes (OR 2.1; 95% CI 1.37-3.32) and age (OR 1.13; 95% CI 1.09-1.14). Despite their young age, almost 20% of HIV-ART treated patients had hypertension, 50% of these were undiagnosed. Blood pressure monitoring is crucial in these patients, especially those identified as high-risk, with prompt life and disability-saving interventions.
Collapse
Affiliation(s)
- Déo Harimenshi
- grid.9966.00000 0001 2165 4861Inserm U1094, IRD U270, CHU Limoges, EpiMaCT-Epidemiology of Chronic Diseases in Tropical Zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, University of Limoges, Limoges, France
| | - Théodore Niyongabo
- grid.7749.d0000 0001 0723 7738Department of Internal Medicine, CHU Kamenge, University of Burundi, Bujumbura, Burundi
| | - Pierre-Marie Preux
- grid.9966.00000 0001 2165 4861Inserm U1094, IRD U270, CHU Limoges, EpiMaCT-Epidemiology of Chronic Diseases in Tropical Zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, University of Limoges, Limoges, France
| | - Victor Aboyans
- grid.9966.00000 0001 2165 4861Inserm U1094, IRD U270, CHU Limoges, EpiMaCT-Epidemiology of Chronic Diseases in Tropical Zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, University of Limoges, Limoges, France ,grid.411178.a0000 0001 1486 4131Department of Cardiology, CHU Limoges, Limoges, France
| | - Ileana Desormais
- grid.9966.00000 0001 2165 4861Inserm U1094, IRD U270, CHU Limoges, EpiMaCT-Epidemiology of Chronic Diseases in Tropical Zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, University of Limoges, Limoges, France ,grid.411178.a0000 0001 1486 4131Department of Vascular Surgery and Vascular Medicine, CHU Limoges, Limoges, France
| |
Collapse
|
128
|
Fadaei R, Davies SS. Oxidative modification of HDL by lipid aldehydes impacts HDL function. Arch Biochem Biophys 2022; 730:109397. [PMID: 36116503 PMCID: PMC9670862 DOI: 10.1016/j.abb.2022.109397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022]
Abstract
Reduced levels of high-density lipoprotein (HDL) cholesterol correlate with increased risk for atherosclerotic cardiovascular diseases and HDL performs functions including reverse cholesterol transport, inhibition of lipid peroxidation, and suppression of inflammation, that would appear critical for cardioprotection. However, several large clinical trials utilizing pharmacologic interventions that elevated HDL cholesterol levels failed to provide cardioprotection to at-risk individuals. The reasons for these unexpected results have only recently begun to be elucidated. HDL cholesterol levels and HDL function can be significantly discordant, so that elevating HDL cholesterol levels may not necessarily lead to increased functional capacity, particularly under conditions that cause HDL to become oxidatively modified, resulting in HDL dysfunction. Here we review evidence that oxidative modifications of HDL, including by reactive lipid aldehydes generated by lipid peroxidation, reduce HDL functionality and that dicarbonyl scavengers that protect HDL against lipid aldehyde modification are beneficial in pre-clinical models of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
129
|
Chen W, Van Beusecum JP, Xiao L, Patrick DM, Ao M, Zhao S, Lopez MG, Billings FT, Cavinato C, Caulk AW, Humphrey JD, Harrison DG. Role of Axl in target organ inflammation and damage due to hypertensive aortic remodeling. Am J Physiol Heart Circ Physiol 2022; 323:H917-H933. [PMID: 36083796 PMCID: PMC9602715 DOI: 10.1152/ajpheart.00253.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
We have shown that excessive endothelial cell stretch causes release of growth arrest-specific 6 (GAS6), which activates the tyrosine kinase receptor Axl on monocytes and promotes immune activation and inflammation. We hypothesized that GAS6/Axl blockade would reduce renal and vascular inflammation and lessen renal dysfunction in the setting of chronic aortic remodeling. We characterized a model of aortic remodeling in mice following a 2-wk infusion of angiotensin II (ANG II). These mice had chronically increased pulse wave velocity, and their aortas demonstrated increased mural collagen. Mechanical testing revealed a marked loss of Windkessel function that persisted for 6 mo following ANG II infusion. Renal function studies showed a reduced ability to excrete a volume load, a progressive increase in albuminuria, and tubular damage as estimated by periodic acid Schiff staining. Treatment with the Axl inhibitor R428 beginning 2 mo after ANG II infusion had a minimal effect on aortic remodeling 2 mo later but reduced the infiltration of T cells, γ/δ T cells, and macrophages into the aorta and kidney and improved renal excretory capacity, reduced albuminuria, and reduced evidence of renal tubular damage. In humans, circulating Axl+/Siglec6+ dendritic cells and phospho-Axl+ cells correlated with pulse wave velocity and aortic compliance measured by transesophageal echo, confirming chronic activation of the GAS6/Axl pathway. We conclude that brief episodes of hypertension induce chronic aortic remodeling, which is associated with persistent low-grade inflammation of the aorta and kidneys and evidence of renal dysfunction. These events are mediated at least in part by GAS6/Axl signaling and are improved with Axl blockade.NEW & NOTEWORTHY In this study, a brief, 2-wk period of hypertension in mice led to progressive aortic remodeling, an increase in pulse wave velocity, and evidence of renal injury, dysfunction, and albuminuria. This end-organ damage was associated with persistent renal and aortic infiltration of CD8+ and γ/δ T cells. We show that this inflammatory response is likely due to GAS6/Axl signaling and can be ameliorated by blocking this pathway. We propose that the altered microvascular mechanical forces caused by increased pulse wave velocity enhance GAS6 release from the endothelium, which in turn activates Axl on myeloid cells, promoting the end-organ damage associated with aortic stiffening.
Collapse
Affiliation(s)
- Wei Chen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin P Van Beusecum
- Ralph H. Johnson Veteran Affairs Medical Center, Charleston, South Carolina
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Liang Xiao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - David M Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Medical Center, Nashville, Tennessee
| | - Mingfang Ao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shilin Zhao
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcos G Lopez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Frederic T Billings
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Alexander W Caulk
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
130
|
Epigenetic Mechanisms Involved in Inflammaging-Associated Hypertension. Curr Hypertens Rep 2022; 24:547-562. [PMID: 35796869 DOI: 10.1007/s11906-022-01214-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the involvement of inflammaging in vascular damage with focus on the epigenetic mechanisms by which inflammaging-induced hypertension is triggered. RECENT FINDINGS Inflammaging in hypertension is a complex condition associated with the production of inflammatory mediators by the immune cells, enhancement of oxidative stress, and tissue remodeling in vascular smooth muscle cells and endothelial cells. Cellular processes are numerous, including inflammasome assembly and cell senescence which may involve mitochondrial dysfunction, autophagy, DNA damage response, dysbiosis, and many others. More recently, a series of noncoding RNAs, mainly microRNAs, have been described as possessing epigenetic actions on the regulation of inflammasome-related hypertension, emerging as a promising therapeutic strategy. Although there are a variety of pharmacological agents that effectively regulate inflammaging-related hypertension, a deeper understanding of the epigenetic events behind the control of vessel deterioration is needed for the treatment or even to prevent the disease onset.
Collapse
|
131
|
Navaneethabalakrishnan S, Smith HL, Arenaz CM, Goodlett BL, McDermott JG, Mitchell BM. Update on Immune Mechanisms in Hypertension. Am J Hypertens 2022; 35:842-851. [PMID: 35704473 PMCID: PMC9527774 DOI: 10.1093/ajh/hpac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/02/2023] Open
Abstract
The contribution of immune cells in the initiation and maintenance of hypertension is undeniable. Several studies have established the association between hypertension, inflammation, and immune cells from the innate and adaptive immune systems. Here, we provide an update to our 2017 American Journal of Hypertension review on the overview of the cellular immune responses involved in hypertension. Further, we discuss the activation of immune cells and their contribution to the pathogenesis of hypertension in different in vivo models. We also highlight existing gaps in the field of hypertension that need attention. The main goal of this review is to provide a knowledge base for translational research to develop therapeutic strategies that can improve cardiovascular health in humans.
Collapse
Affiliation(s)
| | - Hannah L Smith
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Cristina M Arenaz
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Bethany L Goodlett
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Justin G McDermott
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| |
Collapse
|
132
|
Ertuglu LA, Kirabo A. Dendritic Cell Epithelial Sodium Channel in Inflammation, Salt-Sensitive Hypertension, and Kidney Damage. KIDNEY360 2022; 3:1620-1629. [PMID: 36245645 PMCID: PMC9528365 DOI: 10.34067/kid.0001272022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/24/2022] [Indexed: 11/27/2022]
Abstract
Salt-sensitive hypertension is a major risk factor for cardiovascular morbidity and mortality. The pathophysiologic mechanisms leading to different individual BP responses to changes in dietary salt remain elusive. Research in the last two decades revealed that the immune system plays a critical role in the development of hypertension and related end organ damage. Moreover, sodium accumulates nonosmotically in human tissue, including the skin and muscle, shifting the dogma on body sodium balance and its regulation. Emerging evidence suggests that high concentrations of extracellular sodium can directly trigger an inflammatory response in antigen-presenting cells (APCs), leading to hypertension and vascular and renal injury. Importantly, sodium entry into APCs is mediated by the epithelial sodium channel (ENaC). Although the role of the ENaC in renal regulation of sodium excretion and BP is well established, these new findings imply that the ENaC may also exert BP modulatory effects in extrarenal tissue through an immune-dependent pathway. In this review, we discuss the recent advances in our understanding of the pathophysiology of salt-sensitive hypertension with a particular focus on the roles of APCs and the extrarenal ENaC.
Collapse
|
133
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
134
|
Ehret E, Hummler E. Lessons learned about epithelial sodium channels from transgenic mouse models. Curr Opin Nephrol Hypertens 2022; 31:493-501. [PMID: 35894285 PMCID: PMC10022670 DOI: 10.1097/mnh.0000000000000821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review provides an up-to-date understanding about the regulation of epithelial sodium channel (ENaC) expression and function. In particular, we will focus on its implication in renal Na+ and K+ handling and control of blood pressure using transgenic animal models. RECENT FINDINGS In kidney, the highly amiloride-sensitive ENaC maintains whole body Na+ homeostasis by modulating Na+ transport via epithelia. This classical role is mostly confirmed using genetically engineered animal models. Recently identified key signaling pathways that regulate ENaC expression and function unveiled some nonclassical and unexpected channel regulatory processes. If aberrant, these dysregulated mechanisms may also result in the development of salt-dependent hypertension.The purpose of this review is to highlight the most recent findings in renal ENaC regulation and function, in considering data obtained from animal models. SUMMARY Increased ENaC-mediated Na+ transport is a prerequisite for salt-dependent forms of hypertension. To treat salt-sensitive hypertension it is crucial to fully understand the function and regulation of ENaC.
Collapse
Affiliation(s)
- Elodie Ehret
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne
| | - Edith Hummler
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne
- National Center of Competence in Research, Kidney.CH, Zurich, Switzerland
| |
Collapse
|
135
|
Mutengo KH, Masenga SK, Mwesigwa N, Patel KP, Kirabo A. Hypertension and human immunodeficiency virus: A paradigm for epithelial sodium channels? Front Cardiovasc Med 2022; 9:968184. [PMID: 36093171 PMCID: PMC9452753 DOI: 10.3389/fcvm.2022.968184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/10/2022] [Indexed: 02/03/2023] Open
Abstract
Hypertension is a risk factor for end organ damage and death and is more common in persons with HIV compared to the general population. Several mechanisms have been studied in the pathogenesis of hypertension. Current evidence suggests that the epithelial sodium channel (ENaC) plays a key role in regulating blood pressure through the transport of sodium and water across membranes in the kidney tubules, resulting in retention of sodium and water and an altered fluid balance. However, there is scarcity of information that elucidates the role of ENaC in HIV as it relates to increasing the risk for development or pathogenesis of hypertension. This review summarized the evidence to date implicating a potential role for altered ENaC activity in contributing to hypertension in patients with HIV.
Collapse
Affiliation(s)
- Katongo H. Mutengo
- School of Medicine and Health Sciences, HAND Research Group, Mulungushi University, Livingstone Campus, Livingstone, Zambia,School of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Sepiso K. Masenga
- School of Medicine and Health Sciences, HAND Research Group, Mulungushi University, Livingstone Campus, Livingstone, Zambia,School of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine and Dentistry, Kampala International University, Kampala, Uganda
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States,*Correspondence: Annet Kirabo,
| |
Collapse
|
136
|
Abstract
The lymphatic vessels play an essential role in maintaining immune and fluid homeostasis and in the transport of dietary lipids. The discovery of lymphatic endothelial cell-specific markers facilitated the visualization and mechanistic analysis of lymphatic vessels over the past two decades. As a result, lymphatic vessels have emerged as a crucial player in the pathogenesis of several cardiovascular diseases, as demonstrated by worsened disease progression caused by perturbations to lymphatic function. In this review, we discuss the major findings on the role of lymphatic vessels in cardiovascular diseases such as hypertension, obesity, atherosclerosis, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Dakshnapriya Balasubbramanian
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas 77807, USA
| |
Collapse
|
137
|
Lu X, Crowley SD. Actions of Dendritic Cells in the Kidney during Hypertension. Compr Physiol 2022; 12:4087-4101. [PMID: 35950656 DOI: 10.1002/cphy.c210050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The immune response plays a critical role in the pathogenesis of hypertension, and immune cell populations can promote blood pressure elevation via actions in the kidney. Among these cell lineages, dendritic cells (DCs), the most potent antigen-presenting cells, play a central role in regulating immune response during hypertension and kidney disease. DCs have different subtypes, and renal DCs are comprised of the CD103+ CD11b- and CD103- CD11b+ subsets. DCs become mature and express costimulatory molecules on their surface once they encounter antigen. Isolevuglandin-modified proteins function as antigens to activate DCs and trigger them to stimulate T cells. Activated T cells accumulate in the hypertensive kidney, release effector cytokines, promote renal oxidative stress, and promote renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha, interleukin-17A, and interferon-gamma, each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. Fms-like tyrosine kinase 3 ligand-dependent classical DCs are required to sustain the full hypertensive response, but C-X3 -C chemokine receptor 1 positive DCs do not regulate blood pressure. Excess sodium enters the DC through transporters to activate DCs, whereas the ubiquitin editor A20 in dendritic cells constrains blood pressure elevation by limiting T cell activation. By contrast, activation of the salt sensing kinase, serum/glucocorticoid kinase 1 in DCs exacerbates salt-sensitive hypertension. This article discusses recent studies illustrating mechanisms through which DC-T cell interactions modulate levels of pro-hypertensive mediators to regulate blood pressure via actions in the kidney. © 2022 American Physiological Society. Compr Physiol 12:1-15, 2022.
Collapse
Affiliation(s)
- Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| |
Collapse
|
138
|
Pitzer A, Elijovich F, Laffer CL, Ertuglu LA, Sahinoz M, Saleem M, Krishnan J, Dola T, Aden LA, Sheng Q, Raddatz MA, Wanjalla C, Pakala S, Davies SS, Patrick DM, Kon V, Ikizler TA, Kleyman T, Kirabo A. DC ENaC-Dependent Inflammasome Activation Contributes to Salt-Sensitive Hypertension. Circ Res 2022; 131:328-344. [PMID: 35862128 PMCID: PMC9357159 DOI: 10.1161/circresaha.122.320818] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Salt sensitivity of blood pressure is an independent predictor of cardiovascular morbidity and mortality. The exact mechanism by which salt intake increases blood pressure and cardiovascular risk is unknown. We previously found that sodium entry into antigen-presenting cells (APCs) via the amiloride-sensitive epithelial sodium channel EnaC (epithelial sodium channel) leads to the formation of IsoLGs (isolevuglandins) and release of proinflammatory cytokines to activate T cells and modulate salt-sensitive hypertension. In the current study, we hypothesized that ENaC-dependent entry of sodium into APCs activates the NLRP3 (NOD [nucleotide-binding and oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome via IsoLG formation leading to salt-sensitive hypertension. METHODS We performed RNA sequencing on human monocytes treated with elevated sodium in vitro and Cellular Indexing of Transcriptomes and Epitopes by Sequencing analysis of peripheral blood mononuclear cells from participants rigorously phenotyped for salt sensitivity of blood pressure using an established inpatient protocol. To determine mechanisms, we analyzed inflammasome activation in mouse models of deoxycorticosterone acetate salt-induced hypertension as well as salt-sensitive mice with ENaC inhibition or expression, IsoLG scavenging, and adoptive transfer of wild-type dendritic cells into NLRP3 deficient mice. RESULTS We found that high levels of salt exposure upregulates the NLRP3 inflammasome, pyroptotic and apoptotic caspases, and IL (interleukin)-1β transcription in human monocytes. Cellular Indexing of Transcriptomes and Epitopes by Sequencing revealed that components of the NLRP3 inflammasome and activation marker IL-1β dynamically vary with changes in salt loading/depletion. Mechanistically, we found that sodium-induced activation of the NLRP3 inflammasome is ENaC and IsoLG dependent. NLRP3 deficient mice develop a blunted hypertensive response to elevated sodium, and this is restored by the adoptive transfer of NLRP3 replete APCs. CONCLUSIONS These findings reveal a mechanistic link between ENaC, inflammation, and salt-sensitive hypertension involving NLRP3 inflammasome activation in APCs. APC activation via the NLRP3 inflammasome can serve as a potential diagnostic biomarker for salt sensitivity of blood pressure.
Collapse
Affiliation(s)
- Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Fernando Elijovich
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melis Sahinoz
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Jaya Krishnan
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Thanvi Dola
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Luul A Aden
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A. Raddatz
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Celestine Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Suman Pakala
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - David M Patrick
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas Kleyman
- Departments of Medicine, Cell Biology, Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| |
Collapse
|
139
|
Oseghale O, Liong S, Coward-Smith M, To EE, Erlich JR, Luong R, Liong F, Miles M, Norouzi S, Martin C, O’Toole S, Brooks RD, Bozinovski S, Vlahos R, O’Leary JJ, Brooks DA, Selemidis S. Influenza A virus elicits peri-vascular adipose tissue inflammation and vascular dysfunction of the aorta in pregnant mice. PLoS Pathog 2022; 18:e1010703. [PMID: 35930608 PMCID: PMC9385053 DOI: 10.1371/journal.ppat.1010703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/17/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Influenza A virus (IAV) infection during pregnancy initiates significant aortic endothelial and vascular smooth muscle dysfunction, with inflammation and T cell activation, but the details of the mechanism are yet to be clearly defined. Here we demonstrate that IAV disseminates preferentially into the perivascular adipose tissue (PVAT) of the aorta in mice. IAV mRNA levels in the PVAT increased at 1–3 days post infection (d.p.i) with the levels being ~4–8 fold higher compared with the vessel wall. IAV infection also increased Ly6Clow patrolling monocytes and Ly6Chigh pro-inflammatory monocytes in the vessel wall at 3 d.p.i., which was then followed by a greater homing of these monocytes into the PVAT at 6 d.p.i. The vascular immune phenotype was characteristic of a “vascular storm”- like response, with increases in neutrophils, pro-inflammatory cytokines and oxidative stress markers in the PVAT and arterial wall, which was associated with an impairment in endothelium-dependent relaxation to acetylcholine. IAV also triggered a PVAT compartmentalised elevation in CD4+ and CD8+ activated T cells. In conclusion, the PVAT of the aorta is a niche that supports IAV dissemination and a site for perpetuating a profound innate inflammatory and adaptive T cell response. The manifestation of this inflammatory response in the PVAT following IAV infection may be central to the genesis of cardiovascular complications arising during pregnancy. Influenza A virus (IAV) infection remains a major cause of significant disease during pregnancy. IAV infection in pregnancy results in virus dissemination from the lung to the systemic vasculature, thereby initiating profound vascular inflammation and T cell activation that leads to vascular damage. Currently, the details of the mechanism that facilitates this vascular pathology and the influence of IAV dissemination to the vasculature on the perivascular adipose tissue (PVAT) is not clearly defined. Here, we show that IAV disseminates to the PVAT compartment of the vessel at a much larger rate than the vessel wall. We found that IAV infection increased PVAT inflammation characterised by immune cell infiltration, oxidative stress and pro-inflammatory cytokines. This was accompanied by a preferential immune T cell activation in the PVAT. We also found that this vascular inflammatory burden results in vascular endothelial dysfunction that is characterised by an impairment in endothelium dependent relaxation. Our study provides new insights into how IAV utilises the PVAT to promote the vascular inflammatory pathology that disrupts the vasculature in pregnancy and lead to pregnancy complications.
Collapse
Affiliation(s)
- Osezua Oseghale
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Stella Liong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- * E-mail: (SL); (SS)
| | - Madison Coward-Smith
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Eunice E. To
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Jonathan R. Erlich
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Raymond Luong
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Felicia Liong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Mark Miles
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Shaghayegh Norouzi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Cara Martin
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, Dublin, Ireland
- Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
- CERVIVA research consortium, Trinity College Dublin, Dublin, Ireland
| | - Sharon O’Toole
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, Dublin, Ireland
- Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
- CERVIVA research consortium, Trinity College Dublin, Dublin, Ireland
| | - Robert D. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, Dublin, Ireland
- Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
- CERVIVA research consortium, Trinity College Dublin, Dublin, Ireland
| | - Doug A. Brooks
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- * E-mail: (SL); (SS)
| |
Collapse
|
140
|
Krishnan J, de la Visitación N, Hennen EM, Amarnath V, Harrison DG, Patrick DM. IsoLGs (Isolevuglandins) Drive Neutrophil Migration in Hypertension and Are Essential for the Formation of Neutrophil Extracellular Traps. Hypertension 2022; 79:1644-1655. [PMID: 35686559 PMCID: PMC9308685 DOI: 10.1161/hypertensionaha.122.19305] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND IsoLGs (isolevuglandins) are electrophilic products of lipid peroxidation formed in the presence of reactive oxygen species. IsoLGs contribute to hypertension by an unknown mechanism. Studies have shown that reactive oxygen species production drives the formation of neutrophil extracellular traps (NETs) and that NETs accumulate within the aorta and kidneys of patients with hypertension. The purpose of this study was to determine the role of isoLGs in neutrophil migration and NET formation (NETosis) in hypertension. METHODS Mice were treated with Ang II (angiotensin II) and the specific isoLG scavenger 2-hydroxybenzylamine and examined for tissue neutrophil and NET accumulation by single-cell sequencing and flow cytometry. Isolated human neutrophils were studied to determine the role of isoLGs in NETosis and neutrophil chromatin expansion by immunofluorescence and live cell confocal microscopy. RESULTS Single-cell sequencing performed on sham, Ang II, and Ang II+2-hydroxybenzylamine treated mice revealed neutrophils as a primary target of 2-hydroxybenzylamine. Peripheral neutrophil migration, aortic NET accumulation, and renal NET accumulation is blocked with 2-hydroxybenzylamine treatment. In isolated human neutrophils, isoLGs accumulate during NETosis and scavenging of isoLGs prevents NETosis. IsoLGs drive neutrophil chromatin expansion during NETosis and disrupt nucleosome structure. CONCLUSIONS These observations identified a critical role of isoLGs in neutrophil migration and NETosis in hypertension and provide a potential therapy for NET-associated diseases including hypertension and associated end organ damage.
Collapse
Affiliation(s)
- Jaya Krishnan
- Division of Clinical Pharmacology (J.K., N.d.l.V., V.A., D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center
| | - Néstor de la Visitación
- Division of Clinical Pharmacology (J.K., N.d.l.V., V.A., D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center
| | | | - Venkataraman Amarnath
- Division of Clinical Pharmacology (J.K., N.d.l.V., V.A., D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center
| | - David G Harrison
- Division of Clinical Pharmacology (J.K., N.d.l.V., V.A., D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center.,Division of Cardiovascular Medicine (D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center
| | - David M Patrick
- Division of Clinical Pharmacology (J.K., N.d.l.V., V.A., D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center.,Division of Cardiovascular Medicine (D.G.H., D.M.P.), Department of Medicine, Vanderbilt University Medical Center.,Department of Veterans Affairs, Nashville, TN (D.M.P.)
| |
Collapse
|
141
|
Owen A, Patel JM, Parekh D, Bangash MN. Mechanisms of Post-critical Illness Cardiovascular Disease. Front Cardiovasc Med 2022; 9:854421. [PMID: 35911546 PMCID: PMC9334745 DOI: 10.3389/fcvm.2022.854421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged critical care stays commonly follow trauma, severe burn injury, sepsis, ARDS, and complications of major surgery. Although patients leave critical care following homeostatic recovery, significant additional diseases affect these patients during and beyond the convalescent phase. New cardiovascular and renal disease is commonly seen and roughly one third of all deaths in the year following discharge from critical care may come from this cluster of diseases. During prolonged critical care stays, the immunometabolic, inflammatory and neurohumoral response to severe illness in conjunction with resuscitative treatments primes the immune system and parenchymal tissues to develop a long-lived pro-inflammatory and immunosenescent state. This state is perpetuated by persistent Toll-like receptor signaling, free radical mediated isolevuglandin protein adduct formation and presentation by antigen presenting cells, abnormal circulating HDL and LDL isoforms, redox and metabolite mediated epigenetic reprogramming of the innate immune arm (trained immunity), and the development of immunosenescence through T-cell exhaustion/anergy through epigenetic modification of the T-cell genome. Under this state, tissue remodeling in the vascular, cardiac, and renal parenchymal beds occurs through the activation of pro-fibrotic cellular signaling pathways, causing vascular dysfunction and atherosclerosis, adverse cardiac remodeling and dysfunction, and proteinuria and accelerated chronic kidney disease.
Collapse
Affiliation(s)
- Andrew Owen
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jaimin M. Patel
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Mansoor N. Bangash
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Mansoor N. Bangash
| |
Collapse
|
142
|
Rinschen MM, Palygin O, El-Meanawy A, Domingo-Almenara X, Palermo A, Dissanayake LV, Golosova D, Schafroth MA, Guijas C, Demir F, Jaegers J, Gliozzi ML, Xue J, Hoehne M, Benzing T, Kok BP, Saez E, Bleich M, Himmerkus N, Weisz OA, Cravatt BF, Krüger M, Benton HP, Siuzdak G, Staruschenko A. Accelerated lysine metabolism conveys kidney protection in salt-sensitive hypertension. Nat Commun 2022; 13:4099. [PMID: 35835746 PMCID: PMC9283537 DOI: 10.1038/s41467-022-31670-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/27/2022] [Indexed: 01/07/2023] Open
Abstract
Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.
Collapse
Affiliation(s)
- Markus M Rinschen
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- III. Medical Clinic, University Hospital Hamburg Eppendorf, Hamburg, Germany.
- AIAS, Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark.
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xavier Domingo-Almenara
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Omics Sciences Unit, EURECAT, Technology Centre of Catalonia, Reus, Catalonia, Spain
| | - Amelia Palermo
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Fatih Demir
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Megan L Gliozzi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jingchuan Xue
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Martin Hoehne
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Thomas Benzing
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Bernard P Kok
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Markus Bleich
- Institute of Physiology, University Kiel, Kiel, Germany
| | | | - Ora A Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | | | - Marcus Krüger
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - H Paul Benton
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
143
|
Patrick DM, de la Visitación N, Krishnan J, Chen W, Ormseth MJ, Stein CM, Davies SS, Amarnath V, Crofford LJ, Williams JM, Zhao S, Smart CD, Dikalov S, Dikalova A, Xiao L, Van Beusecum JP, Ao M, Fogo AB, Kirabo A, Harrison DG. Isolevuglandins disrupt PU.1-mediated C1q expression and promote autoimmunity and hypertension in systemic lupus erythematosus. JCI Insight 2022; 7:e136678. [PMID: 35608913 PMCID: PMC9310530 DOI: 10.1172/jci.insight.136678] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
We describe a mechanism responsible for systemic lupus erythematosus (SLE). In humans with SLE and in 2 SLE murine models, there was marked enrichment of isolevuglandin-adducted proteins (isoLG adducts) in monocytes and dendritic cells. We found that antibodies formed against isoLG adducts in both SLE-prone mice and humans with SLE. In addition, isoLG ligation of the transcription factor PU.1 at a critical DNA binding site markedly reduced transcription of all C1q subunits. Treatment of SLE-prone mice with the specific isoLG scavenger 2-hydroxybenzylamine (2-HOBA) ameliorated parameters of autoimmunity, including plasma cell expansion, circulating IgG levels, and anti-dsDNA antibody titers. 2-HOBA also lowered blood pressure, attenuated renal injury, and reduced inflammatory gene expression uniquely in C1q-expressing dendritic cells. Thus, isoLG adducts play an essential role in the genesis and maintenance of systemic autoimmunity and hypertension in SLE.
Collapse
Affiliation(s)
- David M. Patrick
- Department of Veterans Affairs, Nashville, Tennessee, USA
- Division of Clinical Pharmacology and
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Néstor de la Visitación
- Division of Clinical Pharmacology and
- Department of Pharmacology, University of Granada, Granada, Spain
| | | | - Wei Chen
- Division of Clinical Pharmacology and
| | - Michelle J. Ormseth
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Rheumatology and Immunology, Department of Medicine, and
| | - C. Michael Stein
- Division of Clinical Pharmacology and
- Division of Rheumatology and Immunology, Department of Medicine, and
| | | | | | | | | | - Shilin Zhao
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Charles D. Smart
- Division of Clinical Pharmacology and
- Department of Molecular Physiology and Biophysics
| | | | | | | | - Justin P. Van Beusecum
- Ralph H. Johnson VA Medical Center and
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Agnes B. Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - David G. Harrison
- Division of Clinical Pharmacology and
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
144
|
Lu X, Crowley SD. The Immune System in Hypertension: a Lost Shaker of Salt 2021 Lewis K. Dahl Memorial Lecture. Hypertension 2022; 79:1339-1347. [PMID: 35545942 DOI: 10.1161/hypertensionaha.122.18554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The seminal observations of Dr Lewis Dahl regarding renal mechanisms of hypertension remain highly relevant in light of more recent experiments showing that immune system dysfunction contributes to hypertension pathogenesis. Dr Dahl established that inappropriate salt retention in the kidney plays a central role via Ohm's Law in permitting blood pressure elevation. Nevertheless, inflammatory cytokines whose expression is induced in the early stages of hypertension can alter renal blood flow and sodium transporter expression and activity to foster renal sodium retention. By elaborating these cytokines and reactive oxygen species, myeloid cells and T lymphocytes can connect systemic inflammatory signals to aberrant kidney functions that allow sustained hypertension. By activating T lymphocytes, antigen-presenting cells such as dendritic cells represent an afferent sensing mechanism triggering T cell activation, cytokine generation, and renal salt and water reabsorption. Manipulating these inflammatory signals to attenuate hypertension without causing prohibitive systemic immunosuppression will pose a challenge, but disrupting actions of inflammatory mediators locally within the kidney may offer a path through which to target immune-mediated mechanisms of hypertension while capitalizing on Dr Dahl's key recognition of the kidney's importance in blood pressure regulation.
Collapse
Affiliation(s)
- Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, NC
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, NC
| |
Collapse
|
145
|
Inflammation, Lymphatics, and Cardiovascular Disease: Amplification by Chronic Kidney Disease. Curr Hypertens Rep 2022; 24:455-463. [PMID: 35727522 DOI: 10.1007/s11906-022-01206-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Kidney disease is a strong modulator of the composition and metabolism of the intestinal microbiome that produces toxins and inflammatory factors. The primary pathways for these harmful factors are blood vessels and nerves. Although lymphatic vessels are responsible for clearance of interstitial fluids, macromolecules, and cells, little is known about whether and how kidney injury impacts the intestinal lymphatic network. RECENT FINDINGS Kidney injury stimulates intestinal lymphangiogenesis, activates lymphatic endothelial cells, and increases mesenteric lymph flow. The mesenteric lymph of kidney-injured animals contains increased levels of cytokines, immune cells, isolevuglandin (IsoLG), a highly reactive dicarbonyl, and of apolipoprotein AI (apoAI). IsoLG is increased in the ileum of kidney injured animals, and intestinal epithelial cells exposed to myeloperoxidase produce more IsoLG. IsoLG-modified apoAI directly increases lymphatic vessel contractions and activates lymphatic endothelial cells. Inhibition of IsoLG by carbonyl scavenger treatment reduces intestinal lymphangiogenesis in kidney-injured animals. Research from our group and others suggests a novel mediator (IsoLG-modified apoAI) and a new pathway (intestinal lymphatic network) in the cross talk between kidneys and intestines and heart. Kidney injury activates intestinal lymphangiogenesis and increases lymphatic flow via mechanisms involving intestinally generated IsoLG. The data identify a new pathway in the kidney gut-heart axis and present a new target for kidney disease-induced intestinal disruptions that may lessen the major adverse consequence of kidney impairment, namely cardiovascular disease.
Collapse
|
146
|
Bencivenga L, De Souto Barreto P, Rolland Y, Hanon O, Vidal JS, Cestac P, Vellas B, Rouch L. Blood pressure variability: a potential marker of aging. Ageing Res Rev 2022; 80:101677. [PMID: 35738476 DOI: 10.1016/j.arr.2022.101677] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/03/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
Abstract
Aging is characterized by alterations in neuro-cardiovascular regulatory mechanisms, leading to impaired physiological variability patterns. Repeated evidence has shown that increased Blood Pressure Variability (BPV) is associated with organ damage and exerts independent predictive value on several health outcomes: cardiovascular events, neurocognitive impairment, metabolic disorders and typical geriatric syndromes such as sarcopenia and frailty. Accordingly, it may constitute the epiphenomenon of the alterations in homeostatic mechanisms, typical of late life. Aging and altered BPV share the same molecular mechanisms, in particular the clinical state of subclinical inflammation has been widely ascertained in advanced age and it is also related to BP dysregulation through altered endothelial function and increased production of ROS. Arterial stiffness and autonomic dysfunction have been associated to impairment in BPV and also represent key features in elderly patients. Furthermore, accumulating evidence in the field of Geroscience has reported that several molecular changes described in cardiovascular aging and altered BPV also relate with the majority of the 9 identified hallmarks of aging. Indeed, BPV may be linked to genomic instability, epigenetic modification and mitochondrial oxidative damage, which represent milestones of aging process. The aim of the present paper is to analyse the interplay between BPV and the pathophysiology of the ageing process, in order to stimulate discussion about the potential role of BPV as a new marker of aging.
Collapse
Affiliation(s)
- Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II"; Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France.
| | - Philipe De Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; UMR INSERM 1295, Université Toulouse III, France
| | - Yves Rolland
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; UMR INSERM 1295, Université Toulouse III, France
| | - Olivier Hanon
- EA4468 Université de Paris, France; Service de gériatrie, Hôpital Broca, AP-HP, Hôpitaux Universitaires Paris Centre, France
| | - Jean-Sébastien Vidal
- EA4468 Université de Paris, France; Service de gériatrie, Hôpital Broca, AP-HP, Hôpitaux Universitaires Paris Centre, France
| | - Philippe Cestac
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; UMR INSERM 1295, Université Toulouse III, France
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; UMR INSERM 1295, Université Toulouse III, France
| | - Laure Rouch
- Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, France; UMR INSERM 1295, Université Toulouse III, France
| |
Collapse
|
147
|
Jia J, Yang JQ, Du YR, Xu Y, Kong D, Zhang XL, Mao JH, Hu GF, Wang KH, Kuang YQ. Transcriptomic Profiling Reveals Underlying Immunoregulation Mechanisms of Resistant Hypertension in Injection Drug Users. J Inflamm Res 2022; 15:3409-3420. [PMID: 35706529 PMCID: PMC9191201 DOI: 10.2147/jir.s361634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/25/2022] [Indexed: 12/18/2022] Open
Abstract
Background Hypertension is a common complication in injection drug users (IDU), especially a high proportion of resistant hypertension occurs among them. However, the involving mechanisms remain largely unknown. Methods We here investigated the key signaling moieties in resistant hypertension in drug users. Analyses were performed with high-throughput transcriptomic sequencing data of peripheral blood from individuals with drug-sensitive hypertension (Ctrl-DS), IDU with resistant hypertension (IDU-DR), and IDU with sensitive hypertension (IDU-DS). Results We showed that 17 and 1 genes in IDU-DS, 48 and 4 genes in IDU-DR were upregulated and downregulated compared Ctrl-DS, and 2 and 4 genes were upregulated and downregulated in IDU-DR compared with IDU-DS, respectively (p ≤ 0.01 and |log2(FC)| ≥ 1). Differentially expressed genes (DEGs) between Ctrl-DS and IDU-DS were mainly involved in Gene ontology terms of immunoglobulin complex and blood microparticle. DEGs between IDU-DS and IDU-DR were mainly involved in immune system process and immunoglobulin complex. DEGs between Ctrl-DS and IDU-DR were mainly involved in immunoglobulin complex, blood microparticle and cytoplasmic vesicle lumen. We identified 2 gene clusters (brown modules, MEbrown; turquoise module, MEturquoise) correlated with IDU-DR and a gene cluster (magenta module, MEmagenta) correlated with IDU-DS by weighted gene co-expression network analysis (WGCNA). Functional analysis demonstrated that pathways of focal adhesion and focalin-1-rich granule lumen were involved in the development of IDU-DR, and the cytosolic large ribosomal subunit may relate to IDU-DR. Further, immune cell infiltration analysis demonstrated that the abundance of dendritic cells (DCs), natural Treg cells (nTreg), and exhausted T cells (Tex) in IDU-DR and IDU-DS, naïve CD8+ T cells in IDU-DS was significantly different compared with that in Ctrl-DS. The abundance of cytotoxic T cells (Tc) was significantly different between IDU-DS and IDU-DR. Conclusion Our findings indicated a potential function of immunoregulation mechanisms for resistant hypertension.
Collapse
Affiliation(s)
- Jie Jia
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, People's Republic of China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Ji-Qun Yang
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, People's Republic of China
| | - Ying-Rong Du
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, People's Republic of China
| | - Yu Xu
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Deshenyue Kong
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, People's Republic of China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Xiu-Ling Zhang
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, People's Republic of China
| | - Jun-Hong Mao
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, People's Republic of China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Gui-Fang Hu
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, People's Republic of China
| | - Kun-Hua Wang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, People's Republic of China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China.,School of Medicine, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yi-Qun Kuang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, People's Republic of China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| |
Collapse
|
148
|
Santos JD, Oliveira Neto JT, Barros PR, Damasceno LEA, Lautherbach N, Assis AP, Silva CAA, Sorgi CA, Faccioli LH, Kettelhut IC, Salgado HC, Carneiro FS, Alves Filho JC, Tostes RC. Th17 cells-linked mechanisms mediate vascular dysfunction induced by testosterone in a mouse model of gender-affirming hormone therapy. Am J Physiol Heart Circ Physiol 2022; 323:H322-H335. [PMID: 35714175 DOI: 10.1152/ajpheart.00182.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clinical data point to adverse cardiovascular events elicited by testosterone replacement therapy. Testosterone is the main hormone used in gender-affirming hormone therapy (GAHT) by transmasculine people. However, the cardiovascular impact of testosterone in experimental models of GAHT remains unknown. Sex hormones modulate T cells activation, and immune mechanisms contribute to cardiovascular risk. The present study evaluated whether testosterone negatively impacts female cardiovascular function by enhancing Th17 cells-linked effector mechanisms. Female (8 weeks-old) C57BL/6J mice received testosterone (48 mg.Kg-1.week-1) for 8 weeks. Male mice were used for phenotypical comparisons. The hormone-treatment in female mice increased circulating testosterone to levels observed in male mice. Testosterone increased lean body mass and body mass index, and decreased perigonadal fat mass, mimicking clinical findings. After 8 weeks, testosterone decreased endothelium-dependent vasodilation and increased circulating Th17 cells. After 24 weeks, testosterone increased blood pressure in female mice. Ovariectomy did not intensify phenotypical or cardiovascular effects by testosterone. Female mice lacking T and B cells [Rag1 knockout (-/-)], as well as female mice lacking IL-17 receptor (IL-17Ra-/-), did not exhibit vascular dysfunction induced by testosterone. Testosterone impaired endothelium-dependent vasodilation in female mice lacking γδ T cells, similarly to the observed in wild type female mice. Adoptive transfer of CD4+ T cells restored testosterone-induced vascular dysfunction in Rag1-/- female mice. Together, these data suggest that CD4+ T cells, most likely Th17 cells, are central to vascular dysfunction induced by testosterone in female mice, indicating that changes in immune cells balance are important in the GAHT in transmasculine people.
Collapse
Affiliation(s)
- Jeimison D Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - José T Oliveira Neto
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Paula R Barros
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Natalia Lautherbach
- Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Ana P Assis
- Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Carlos A A Silva
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Carlos A Sorgi
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters at Ribeira Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Lucia H Faccioli
- Department of Clinical Analyses, Toxicology and Food Science, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Isis C Kettelhut
- Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Helio C Salgado
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Jose C Alves Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
149
|
Parsa D, Aden LA, Pitzer A, Ding T, Yu C, Diedrich A, Milne GL, Kirabo A, Shibao CA. Enhanced parasympathetic cholinergic activity with galantamine inhibited lipid-induced oxidative stress in obese African Americans. Mol Med 2022; 28:60. [PMID: 35659521 PMCID: PMC9164360 DOI: 10.1186/s10020-022-00486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND African Americans (AAs) are disproportionately affected by cardiovascular disease (CVD), they are 20% more likely to die from CVD than whites, chronic exposure to inflammation and oxidative stress contributes to CVD. In previous studies, enhancing parasympathetic cholinergic activity has been shown to decrease inflammation. Considering that AAs have decreased parasympathetic activity compared to whites, we hypothesize that stimulating it with a central acetylcholinesterase (AChE) inhibitor, galantamine, would prevent lipid-induced oxidative stress. OBJECTIVE To test the hypothesis that acute dose of galantamine, an AChE inhibitor, decreases lipid-induced oxidative stress in obese AAs. METHODS Proof-of-concept, double-blind, randomized, placebo-controlled, crossover study that tested the effect of a single dose of 16 mg of galantamine versus placebo on lipid-induced oxidative stress in obese AAs. Subjects were studied on two separate days, one week apart. In each study day, 16 mg or matching placebo was administered before 20% intralipids infusion at doses of 0.8 mL/m2/min with heparin at doses of 200 U/h for 4 h. Outcomes were assessed at baseline, 2 and 4 h during the infusion. MAIN OUTCOME MEASURES Changes in F2-isoprostane (F2-IsoPs), marker of oxidative stress, measured in peripheral blood mononuclear cells (PBMC) and in plasma at baseline, 2, and 4-h post-lipid infusion. Secondary outcomes include changes in inflammatory cytokines (IL-6, TNF alpha). RESULTS A total of 32 obese AA women were screened and fourteen completed the study (age 37.8 ± 10.70 years old, BMI 38.7 ± 3.40 kg/m2). Compared to placebo, 16 mg of galantamine significantly inhibited the increase in F2-IsoPs in PBMC (0.007 ± 0.008 vs. - 0.002 ± 0.006 ng/sample, P = 0.016), and plasma (0.01 ± 0.02 vs. - 0.003 ± 0.01 ng/mL, P = 0.023). Galantamine also decreased IL-6 (11.4 ± 18.45 vs. 7.7 ± 15.10 pg/mL, P = 0.021) and TNFα levels (18.6 ± 16.33 vs. 12.9 ± 6.16 pg/mL, P = 0.021, 4-h post lipid infusion) compared with placebo. These changes were associated with an increased plasma acetylcholine levels induced by galantamine (50.5 ± 10.49 vs. 43.6 ± 13.38 during placebo pg/uL, P = 0.025). CONCLUSIONS In this pilot, proof-of-concept study, enhancing parasympathetic nervous system (PNS) cholinergic activity with galantamine inhibited lipid-induced oxidative stress and inflammation induced by lipid infusion in obese AAs. TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT02365285.
Collapse
Affiliation(s)
- Dena Parsa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA
| | - Luul A Aden
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA
| | - Ashley Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA
| | - Tan Ding
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andre Diedrich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Cyndya A Shibao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 506 Robinson Research Building, Nashville, TN, 37232, USA.
| |
Collapse
|
150
|
Su S, Chen R, Zhang S, Shu H, Luo J. Immune system changes in those with hypertension when infected with SARS-CoV-2. Cell Immunol 2022; 378:104562. [PMID: 35901625 PMCID: PMC9183242 DOI: 10.1016/j.cellimm.2022.104562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) outbreak has become an evolving global health crisis. With an increasing incidence of primary hypertension, there is greater awareness of the relationship between primary hypertension and the immune system [including CD4+, CD8+ T cells, interleukin-17 (IL-17)/T regulatory cells (Treg) balance, macrophages, natural killer (NK) cells, neutrophils, B cells, and cytokines]. Hypertension is associated with an increased risk of various infections, post-infection complications, and increased mortality from severe infections. Despite ongoing reports on the epidemiological and clinical features of COVID-19, no articles have systematically addressed the role of primary hypertension in COVID-19 or how COVID-19 affects hypertension or specific treatment in these high-risk groups. Here, we synthesize recent advances in understanding the relationship between primary hypertension and COVID-19 and its underlying mechanisms and provide specific treatment guidelines for these high-risk groups.
Collapse
|