Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Obesity, kidney dysfunction and hypertension: mechanistic links

Abstract

Excessive adiposity raises blood pressure and accounts for 65–75% of primary hypertension, which is a major driver of cardiovascular and kidney diseases. In obesity, abnormal kidney function and associated increases in tubular sodium reabsorption initiate hypertension, which is often mild before the development of target organ injury. Factors that contribute to increased sodium reabsorption in obesity include kidney compression by visceral, perirenal and renal sinus fat; increased renal sympathetic nerve activity (RSNA); increased levels of anti-natriuretic hormones, such as angiotensin II and aldosterone; and adipokines, particularly leptin. The renal and neurohormonal pathways of obesity and hypertension are intertwined. For example, leptin increases RSNA by stimulating the central nervous system proopiomelanocortin–melanocortin 4 receptor pathway, and kidney compression and RSNA contribute to renin–angiotensin–aldosterone system activation. Glucocorticoids and/or oxidative stress may also contribute to mineralocorticoid receptor activation in obesity. Prolonged obesity and progressive renal injury often lead to the development of treatment-resistant hypertension. Patient management therefore often requires multiple antihypertensive drugs and concurrent treatment of dyslipidaemia, insulin resistance, diabetes and inflammation. If more effective strategies for the prevention and control of obesity are not developed, cardiorenal, metabolic and other obesity-associated diseases could overwhelm health-care systems in the future.

Key points

  • Obesity is associated with chronic diseases, including hypertension, which is a major risk factor for chronic kidney disease and cardiovascular diseases such as stroke, myocardial infarction and heart failure.

  • Excessive weight gain, especially when associated with visceral obesity, raises blood pressure and is the most important known risk factor for primary (essential) hypertension.

  • Abnormal kidney function, which is associated with increased tubular sodium reabsorption, has a key role in initiating obesity-associated hypertension.

  • Mechanisms that initiate obesity-induced sodium retention include kidney compression by visceral, perirenal and renal sinus fat, stimulation of the renin–angiotensin–aldosterone system, aldosterone-independent mineralocorticoid receptor activation and activation of the sympathetic nervous system.

  • Sympathetic activation in obesity may be mediated by hypoxia, chemoreceptor activation, baroreflex dysfunction and adipokines, including leptin, which activates the central nervous system melanocortin pathway.

  • Chronic obesity may gradually amplify hypertension, resulting in resistance to antihypertensive treatment and initiating a pathophysiological cascade of factors that exacerbate target organ injury.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Obesity shifts the frequency distribution of blood pressure.
Fig. 2: Potential effects of kidney compression on renal haemodynamics, sodium reabsorption and renin secretion.
Fig. 3: Mechanisms of obesity-induced hypertension, renal injury and cardiovascular disease.
Fig. 4: Potential mechanisms and consequences of MR activation in obesity.
Fig. 5: Effects of CNS leptin–melanocortin activation on blood pressure and metabolic functions.
Fig. 6: Obesity-induced hypertension and the effects of renal denervation.
Fig. 7: Potential mechanisms of SNS activation in obesity.

Similar content being viewed by others

References

  1. NCD Risk Factor Collaboration. Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet 390, 2627–2642 (2017).

    Article  Google Scholar 

  2. Afshin, A., Reitsma, M. B. & Murray, C. J. L. Health effects of overweight and obesity in 195 countries. N. Engl. J. Med. 377, 1496–1497 (2017).

    PubMed  Google Scholar 

  3. Gregg, E. W. & Shaw, J. E. Global health effects of overweight and obesity. N. Engl. J. Med. 377, 80–81 (2017).

    Article  PubMed  Google Scholar 

  4. World Health Organization. Obesity and overweight — key facts. WHO http://www.who.int/mediacentre/factsheets/fs311/en/ (2018).

  5. Benjamin, E. J. et al. Heart disease and stroke statistics-2018 update: a report from the American Heart Association. Circulation 137, e67–e492 (2018).

    Article  PubMed  Google Scholar 

  6. Hall, J. E., do Carmo, J. M., da Silva, A. A., Wang, Z. & Hall, M. E. Obesity-induced hypertension: interaction of neurohumoral and renal mechanisms. Circ. Res. 116, 991–1006 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Garrison, R. J., Kannel, W. B., Stokes, J. III & Castelli, W. P. Incidence and precursors of hypertension in young adults: the Framingham Offspring Study. Prev. Med. 16, 235–251 (1987).

    Article  CAS  PubMed  Google Scholar 

  8. USRDS Coordinating Center. United States Renal Data System Annual Report. USRDS https://www.usrds.org/adr.aspx (2017).

  9. Hall, J. E. et al. Is obesity a major cause of chronic kidney disease? Adv. Ren. Replace. Ther. 11, 41–54 (2004).

    Article  PubMed  Google Scholar 

  10. Hall, M. E. et al. Obesity, hypertension, and chronic kidney disease. Int. J. Nephrol. Renovasc. Dis. 7, 75–88 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Whaley-Connell, A. & Sowers, J. R. Obesity and kidney disease: from population to basic science and the search for new therapeutic targets. Kidney Int. 92, 313–323 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. DeMarco, V. G., Aroor, A. R. & Sowers, J. R. The pathophysiology of hypertension in patients with obesity. Nat. Rev. Endocrinol. 10, 364–376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Neeland, I. J., Poirier, P. & Despres, J. P. Cardiovascular and metabolic heterogeneity of obesity: clinical challenges and implications for management. Circulation 137, 1391–1406 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Tchernof, A. & Despres, J. P. Pathophysiology of human visceral obesity: an update. Physiol. Rev. 93, 359–404 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Mohammed, B. S., Cohen, S., Reeds, D., Young, V. L. & Klein, S. Long-term effects of large-volume liposuction on metabolic risk factors for coronary heart disease. Obesity 16, 2648–2651 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Schiavon, C. A. et al. Effects of bariatric surgery in obese patients with hypertension: the GATEWAY randomized trial (gastric bypass to treat obese patients with steady hypertension). Circulation 137, 1132–1142 (2018).

    Article  PubMed  Google Scholar 

  17. Schauer, P. R. et al. Bariatric surgery versus intensive medical therapy for diabetes — 5-year outcomes. N. Engl. J. Med. 376, 641–651 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Piche, M. E., Poirier, P., Lemieux, I. & Despres, J. P. Overview of epidemiology and contribution of obesity and body fat distribution to cardiovascular disease: an update. Prog. Cardiovasc. Dis. 61, 103–113 (2018).

    Article  PubMed  Google Scholar 

  19. Chughtai, H. L. et al. Renal sinus fat and poor blood pressure control in middle-aged and elderly individuals at risk for cardiovascular events. Hypertension 56, 901–906 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Foster, M. C. et al. Fatty kidney, hypertension, and chronic kidney disease: the Framingham Heart Study. Hypertension 58, 784–790 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Chandra, A. et al. The relationship of body mass and fat distribution with incident hypertension: observations from the dallas heart study. J. Am. Coll. Cardiol. 64, 997–1002 (2014).

    Article  PubMed  Google Scholar 

  22. Ortega, F. B., Lavie, C. J. & Blair, S. N. Obesity and cardiovascular disease. Circ. Res. 118, 1752–1770 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Cui, Z., Truesdale, K. P., Bradshaw, P. T., Cai, J. & Stevens, J. Three-year weight change and cardiometabolic risk factors in obese and normal weight adults who are metabolically healthy: the atherosclerosis risk in communities study. Int. J. Obes. 39, 1203–1208 (2015).

    Article  CAS  Google Scholar 

  24. Eckel, N. et al. Transition from metabolic healthy to unhealthy phenotypes and association with cardiovascular disease risk across BMI categories in 90 257 women (the Nurses’ Health Study): 30 year follow-up from a prospective cohort study. Lancet Diabetes Endocrinol. 6, 714–724 (2018).

    Article  PubMed  Google Scholar 

  25. Jones, D. W., Kim, J. S., Andrew, M. E., Kim, S. J. & Hong, Y. P. Body mass index and blood pressure in Korean men and women: the Korean National Blood Pressure Survey. J. Hypertens. 12, 1433–1437 (1994).

    Article  CAS  PubMed  Google Scholar 

  26. Bramlage, P. et al. Hypertension in overweight and obese primary care patients is highly prevalent and poorly controlled. Am. J. Hypertens. 17, 904–910 (2004).

    Article  PubMed  Google Scholar 

  27. Whelton, P. K. et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 71, 1269–1324 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Nyamdorj, R. et al. Comparison of body mass index with waist circumference, waist-to-hip ratio, and waist-to-stature ratio as a predictor of hypertension incidence in Mauritius. J. Hypertens. 26, 866–870 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Field, A. E. et al. Impact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch. Intern. Med. 161, 1581–1586 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Shihab, H. M. et al. Body mass index and risk of incident hypertension over the life course: the Johns Hopkins Precursors Study. Circulation 126, 2983–2989 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Harrap, S. B. & Charchar, F. J. Genetics of blood pressure: time to curate the collection. J. Hypertens. 35, 1360–1362 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Saad, M. F. et al. Racial differences in the relation between blood pressure and insulin resistance. N. Engl. J. Med. 324, 733–739 (1991).

    Article  CAS  PubMed  Google Scholar 

  33. Weyer, C. et al. Ethnic differences in insulinemia and sympathetic tone as links between obesity and blood pressure. Hypertension 36, 531–537 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. de Courten, M. P., Pettitt, D. J. & Knowler, W. C. Hypertension in Pima Indians: prevalence and predictors. Public Health Rep. 111 (Suppl. 2), 40–43 (1996).

    Google Scholar 

  35. Hall, J. E., Crook, E. D., Jones, D. W., Wofford, M. R. & Dubbert, P. M. Mechanisms of obesity-associated cardiovascular and renal disease. Am. J. Med. Sci. 324, 127–137 (2002).

    Article  PubMed  Google Scholar 

  36. Alpert, M. A., Karthikeyan, K., Abdullah, O. & Ghadban, R. Obesity and cardiac remodeling in adults: mechanisms and clinical implications. Prog. Cardiovasc. Dis. 61, 114–123 (2018).

    Article  PubMed  Google Scholar 

  37. Van Vliet, B. N., Hall, J. E., Mizelle, H. L., Montani, J. P. & Smith, M. J. Jr. Reduced parasympathetic control of heart rate in obese dogs. Am. J. Physiol. 269, H629–H637 (1995).

    PubMed  Google Scholar 

  38. Raitakari, M. et al. Weight reduction with very-low-caloric diet and endothelial function in overweight adults: role of plasma glucose. Arterioscler. Thromb. Vasc. Biol. 24, 124–128 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Wasserman, D. H., Wang, T. J. & Brown, N. J. The vasculature in prediabetes. Circ. Res. 122, 1135–1150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wildman, R. P., Mackey, R. H., Bostom, A., Thompson, T. & Sutton-Tyrrell, K. Measures of obesity are associated with vascular stiffness in young and older adults. Hypertension 42, 468–473 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Lavie, C. J., Arena, R., Alpert, M. A., Milani, R. V. & Ventura, H. O. Management of cardiovascular diseases in patients with obesity. Nat. Rev. Cardiol. 15, 45–56 (2018).

    Article  PubMed  Google Scholar 

  42. Tonneijck, L. et al. Glomerular hyperfiltration in diabetes: mechanisms, clinical significance, and treatment. J. Am. Soc. Nephrol. 28, 1023–1039 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Carlstrom, M., Wilcox, C. S. & Arendshorst, W. J. Renal autoregulation in health and disease. Physiol. Rev. 95, 405–511 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Woods, L. L., Mizelle, H. L. & Hall, J. E. Control of renal hemodynamics in hyperglycemia: possible role of tubuloglomerular feedback. Am. J. Physiol. 252, F65–F73 (1987).

    CAS  PubMed  Google Scholar 

  45. Hall, J. E. et al. Obesity-associated hypertension and kidney disease. Curr. Opin. Nephrol. Hypertens. 12, 195–200 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Woods, L. L., Mizelle, H. L., Montani, J. P. & Hall, J. E. Mechanisms controlling renal hemodynamics and electrolyte excretion during amino acids. Am. J. Physiol. 251, F303–F312 (1986).

    CAS  PubMed  Google Scholar 

  47. de Paula, R. B., da Silva, A. A. & Hall, J. E. Aldosterone antagonism attenuates obesity-induced hypertension and glomerular hyperfiltration. Hypertension 43, 41–47 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Hall, J. E., Granger, J. P., Smith, M. J. Jr & Premen, A. J. Role of renal hemodynamics and arterial pressure in aldosterone “escape”. Hypertension 6, I183–I192 (1984).

    Article  CAS  PubMed  Google Scholar 

  49. Fu, Y. et al. Aldosterone blunts tubuloglomerular feedback by activating macula densa mineralocorticoid receptors. Hypertension 59, 599–606 (2012).

    Article  CAS  PubMed  Google Scholar 

  50. Granger, J. P. et al. Role of nitric oxide in modulating renal function and arterial pressure during chronic aldosterone excess. Am. J. Physiol. 276, R197–R202 (1999).

    CAS  PubMed  Google Scholar 

  51. Hall, J. E. Renal dysfunction, rather than nonrenal vascular dysfunction, mediates salt-induced hypertension. Circulation 133, 894–906 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Sugerman, H., Windsor, A., Bessos, M. & Wolfe, L. Intra-abdominal pressure, sagittal abdominal diameter and obesity comorbidity. J. Intern. Med. 241, 71–79 (1997).

    Article  CAS  PubMed  Google Scholar 

  53. Hall, J. E. Louis, K. Dahl Memorial Lecture. Renal and cardiovascular mechanisms of hypertension in obesity. Hypertension 23, 381–394 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Alonso-Galicia, M., Dwyer, T. M., Herrera, G. A. & Hall, J. E. Increased hyaluronic acid in the inner renal medulla of obese dogs. Hypertension 25, 888–892 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Dwyer, T. M. et al. Distribution of renal medullary hyaluronan in lean and obese rabbits. Kidney Int. 58, 721–729 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Hall, J. E., Brands, M. W. & Henegar, J. R. Mechanisms of hypertension and kidney disease in obesity. Ann. NY Acad. Sci. 892, 91–107 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Adeosun, S. O. et al. Loss of biliverdin reductase-A promotes lipid accumulation and lipotoxicity in mouse proximal tubule cells. Am. J. Physiol. Renal Physiol. 315, F323–F331 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Unger, R. H. & Scherer, P. E. Gluttony, sloth and the metabolic syndrome: a roadmap to lipotoxicity. Trends Endocrinol. Metab. 21, 345–352 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Engeli, S. & Sharma, A. M. The renin-angiotensin system and natriuretic peptides in obesity-associated hypertension. J. Mol. Med. 79, 21–29 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Cabandugama, P. K., Gardner, M. J. & Sowers, J. R. The renin angiotensin aldosterone system in obesity and hypertension: roles in the cardiorenal metabolic syndrome. Med. Clin. North Am. 101, 129–137 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Schutten, M. T., Houben, A. J., de Leeuw, P. W. & Stehouwer, C. D. The link between adipose tissue renin-angiotensin-aldosterone system signaling and obesity-associated hypertension. Physiology 32, 197–209 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Alonso-Galicia, M., Brands, M. W., Zappe, D. H. & Hall, J. E. Hypertension in obese Zucker rats. Role of angiotensin II and adrenergic activity. Hypertension 28, 1047–1054 (1996).

    Article  CAS  PubMed  Google Scholar 

  63. Hall, J. E. et al. Hypertension: physiology and pathophysiology. Compr. Physiol. 2, 2393–2442 (2012).

    PubMed  Google Scholar 

  64. Dorresteijn, J. A. et al. Differential effects of renin-angiotensin-aldosterone system inhibition, sympathoinhibition and diuretic therapy on endothelial function and blood pressure in obesity-related hypertension: a double-blind, placebo-controlled cross-over trial. J. Hypertens. 31, 393–403 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Grassi, G. et al. Comparative effects of candesartan and hydrochlorothiazide on blood pressure, insulin sensitivity, and sympathetic drive in obese hypertensive individuals: results of the CROSS study. J. Hypertens. 21, 1761–1769 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Reisin, E. et al. Lisinopril versus hydrochlorothiazide in obese hypertensive patients: a multicenter placebo-controlled trial. Treatment in Obese Patients With Hypertension (TROPHY) study group. Hypertension 30, 140–145 (1997).

    Article  CAS  PubMed  Google Scholar 

  67. Brenner, B. M. et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N. Engl. J. Med. 345, 861–869 (2001).

    Article  CAS  PubMed  Google Scholar 

  68. Lewis, E. J. et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N. Engl. J. Med. 345, 851–860 (2001).

    Article  CAS  PubMed  Google Scholar 

  69. Dinh Cat, A. N., Friederich-Persson, M., White, A. & Touyz, R. M. Adipocytes, aldosterone and obesity-related hypertension. J. Mol. Endocrinol. 57, F7–F21 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Davel, A. P., Jaffe, I. Z., Tostes, R. C., Jaisser, F. & Belin de Chantemele, E. J. New roles of aldosterone and mineralocorticoid receptors in cardiovascular disease: translational and sex-specific effects. Am. J. Physiol. Heart Circ. Physiol. 315, H989–H999 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Faulkner, J. L., Bruder-Nascimento, T. & Belin de Chantemele, E. J. The regulation of aldosterone secretion by leptin: implications in obesity-related cardiovascular disease. Curr. Opin. Nephrol. Hypertens. 27, 63–69 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bomback, A. S., Muskala, P., Bald, E., Chwatko, G. & Nowicki, M. Low-dose spironolactone, added to long-term ACE inhibitor therapy, reduces blood pressure and urinary albumin excretion in obese patients with hypertensive target organ damage. Clin. Nephrol. 72, 449–456 (2009).

    Article  CAS  PubMed  Google Scholar 

  73. Dudenbostel, T. & Calhoun, D. A. Use of aldosterone antagonists for treatment of uncontrolled resistant hypertension. Am. J. Hypertens. 30, 103–109 (2017).

    Article  CAS  PubMed  Google Scholar 

  74. de Souza, F., Muxfeldt, E., Fiszman, R. & Salles, G. Efficacy of spironolactone therapy in patients with true resistant hypertension. Hypertension 55, 147–152 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Hwang, M. H. et al. Mineralocorticoid receptors modulate vascular endothelial function in human obesity. Clin. Sci. 125, 513–520 (2013).

    Article  CAS  Google Scholar 

  76. Kosmala, W., Przewlocka-Kosmala, M., Szczepanik-Osadnik, H., Mysiak, A. & Marwick, T. H. Fibrosis and cardiac function in obesity: a randomised controlled trial of aldosterone blockade. Heart 99, 320–326 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Andersen, K. Aldosterone synthase inhibition in hypertension. Curr. Hypertens. Rep. 15, 484–488 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Namsolleck, P. & Unger, T. Aldosterone synthase inhibitors in cardiovascular and renal diseases. Nephrol. Dial. Transplant. 29, i62–i68 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Calhoun, D. A. et al. Effects of a novel aldosterone synthase inhibitor for treatment of primary hypertension: results of a randomized, double-blind, placebo- and active-controlled phase 2 trial. Circulation 124, 1945–1955 (2011).

    Article  CAS  PubMed  Google Scholar 

  80. Karns, A. D., Bral, J. M., Hartman, D., Peppard, T. & Schumacher, C. Study of aldosterone synthase inhibition as an add-on therapy in resistant hypertension. J. Clin. Hypertens. 15, 186–192 (2013).

    Article  CAS  Google Scholar 

  81. Liao, W. H. et al. Aldosterone deficiency in mice burdens respiration and accentuates diet-induced hyperinsulinemia and obesity. JCI Insight 3, 99015 (2018).

    Article  PubMed  Google Scholar 

  82. Fujita, T. Mechanism of salt-sensitive hypertension: focus on adrenal and sympathetic nervous systems. J. Am. Soc. Nephrol. 25, 1148–1155 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Chapman, K., Holmes, M. & Seckl, J. 11beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. Physiol. Rev. 93, 1139–1206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Funder, J. W. Apparent mineralocorticoid excess. J. Steroid Biochem. Mol. Biol. 165, 151–153 (2017).

    Article  CAS  PubMed  Google Scholar 

  85. Williams, B. et al. Endocrine and haemodynamic changes in resistant hypertension, and blood pressure responses to spironolactone or amiloride: the PATHWAY-2 mechanisms substudies. Lancet Diabetes Endocrinol. 6, 464–475 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Williams, B. et al. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet 386, 2059–2068 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Carey, R. M. et al. Resistant hypertension: detection, evaluation, and management: a scientific statement from the American Heart Association. Hypertension 72, e53–e90 (2018).

    Article  CAS  PubMed  Google Scholar 

  88. Calhoun, D. A. Fluid retention, aldosterone excess, and treatment of resistant hypertension. Lancet Diabetes Endocrinol. 6, 431–433 (2018).

    Article  PubMed  Google Scholar 

  89. Jordan, J., Birkenfeld, A. L., Melander, O. & Moro, C. Natriuretic peptides in cardiovascular and metabolic crosstalk: implications for hypertension management. Hypertension 72, 270–276 (2018).

    Article  CAS  PubMed  Google Scholar 

  90. Asferg, C. L. et al. Relative atrial natriuretic peptide deficiency and inadequate renin and angiotensin II suppression in obese hypertensive men. Hypertension 62, 147–153 (2013).

    Article  CAS  PubMed  Google Scholar 

  91. Asferg, C. L., Andersen, U. B., Linneberg, A., Goetze, J. P. & Jeppesen, J. L. Obese hypertensive men have lower circulating proatrial natriuretic peptide concentrations despite greater left atrial size. Am. J. Hypertens. 31, 645–650 (2018).

    Article  PubMed  Google Scholar 

  92. Savoia, C., Volpe, M., Alonzo, A., Rossi, C. & Rubattu, S. Natriuretic peptides and cardiovascular damage in the metabolic syndrome: molecular mechanisms and clinical implications. Clin. Sci. 118, 231–240 (2010).

    Article  Google Scholar 

  93. Wang, T. J. et al. Impact of obesity on plasma natriuretic peptide levels. Circulation 109, 594–600 (2004).

    Article  CAS  PubMed  Google Scholar 

  94. Grandi, A. M. et al. Natriuretic peptides as markers of preclinical cardiac disease in obesity. Eur. J. Clin. Invest. 34, 342–348 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. Abdulle, A. M. et al. Plasma N terminal pro-brain natriuretic peptide levels and its determinants in a multi-ethnic population. J. Hum. Hypertens. 21, 647–653 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Birkenfeld, A. L. et al. Lipid mobilization with physiological atrial natriuretic peptide concentrations in humans. J. Clin. Endocrinol. Metab. 90, 3622–3628 (2005).

    Article  CAS  PubMed  Google Scholar 

  97. Birkenfeld, A. L. et al. Atrial natriuretic peptide induces postprandial lipid oxidation in humans. Diabetes 57, 3199–3204 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Miyashita, K. et al. Natriuretic peptides/cGMP/cGMP-dependent protein kinase cascades promote muscle mitochondrial biogenesis and prevent obesity. Diabetes 58, 2880–2892 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Bordicchia, M. et al. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J. Clin. Invest. 122, 1022–1036 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Canfora, E. E., Meex, R. C. R., Venema, K. & Blaak, E. E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. https://doi.org/10.1038/s41574-019-0156-z (2019).

    Article  PubMed  Google Scholar 

  101. Aydin, O., Nieuwdorp, M. & Gerdes, V. The gut microbiome as a target for the treatment of type 2 diabetes. Curr. Diab Rep. 18, 55 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ridaura, V. K. et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341, 1241214 (2013).

    Article  CAS  PubMed  Google Scholar 

  103. Yang, T., Richards, E. M., Pepine, C. J. & Raizada, M. K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 14, 442–456 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wilck, N. et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature 551, 585–589 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Santisteban, M. M. et al. Hypertension-linked pathophysiological alterations in the gut. Circ. Res. 120, 312–323 (2017).

    Article  CAS  PubMed  Google Scholar 

  106. Riedl, R. A., Atkinson, S. N., Burnett, C. M. L., Grobe, J. L. & Kirby, J. R. The gut microbiome, energy homeostasis, and implications for hypertension. Curr. Hypertens. Rep. 19, 27 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Richards, E. M., Pepine, C. J., Raizada, M. K. & Kim, S. The gut, its microbiome, and hypertension. Curr. Hypertens. Rep. 19, 36 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Afsar, B., Vaziri, N. D., Aslan, G., Tarim, K. & Kanbay, M. Gut hormones and gut microbiota: implications for kidney function and hypertension. J. Am. Soc. Hypertens. 10, 954–961 (2016).

    Article  CAS  PubMed  Google Scholar 

  109. Muskiet, M. H. A. et al. GLP-1 and the kidney: from physiology to pharmacology and outcomes in diabetes. Nat. Rev. Nephrol. 13, 605–628 (2017).

    Article  CAS  PubMed  Google Scholar 

  110. Zhu, Q. & Scherer, P. E. Immunologic and endocrine functions of adipose tissue: implications for kidney disease. Nat. Rev. Nephrol. 14, 105–120 (2018).

    Article  CAS  PubMed  Google Scholar 

  111. Abraham, T. M., Pedley, A., Massaro, J. M., Hoffmann, U. & Fox, C. S. Association between visceral and subcutaneous adipose depots and incident cardiovascular disease risk factors. Circulation 132, 1639–1647 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. D’Souza, K., Nzirorera, C. & Kienesberger, P. C. Lipid metabolism and signaling in cardiac lipotoxicity. Biochim. Biophys. Acta 1861, 1513–1524 (2016).

    Article  CAS  PubMed  Google Scholar 

  113. Yamauchi, T. et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med. 8, 1288–1295 (2002).

    Article  CAS  PubMed  Google Scholar 

  114. Ohashi, K., Ouchi, N. & Matsuzawa, Y. Adiponectin and hypertension. Am. J. Hypertens. 24, 263–269 (2011).

    Article  CAS  PubMed  Google Scholar 

  115. Wang, Z. V. & Scherer, P. E. Adiponectin, cardiovascular function, and hypertension. Hypertension 51, 8–14 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Sabbatini, A. R., Fontana, V., Laurent, S. & Moreno, H. An update on the role of adipokines in arterial stiffness and hypertension. J. Hypertens. 33, 435–444 (2015).

    Article  CAS  PubMed  Google Scholar 

  117. Ohashi, K. et al. Adiponectin replenishment ameliorates obesity-related hypertension. Hypertension 47, 1108–1116 (2006).

    Article  CAS  PubMed  Google Scholar 

  118. Kim, D. H., Kim, C., Ding, E. L., Townsend, M. K. & Lipsitz, L. A. Adiponectin levels and the risk of hypertension: a systematic review and meta-analysis. Hypertension 62, 27–32 (2013).

    Article  CAS  PubMed  Google Scholar 

  119. Pan, W. W. & Myers, M. G. Jr. Leptin and the maintenance of elevated body weight. Nat. Rev. Neurosci. 19, 95–105 (2018).

    Article  CAS  PubMed  Google Scholar 

  120. Schwartz, M. W. et al. Central nervous system control of food intake. Nature 404, 661–671 (2000).

    Article  CAS  PubMed  Google Scholar 

  121. Hall, J. E. et al. Obesity-induced hypertension: role of sympathetic nervous system, leptin, and melanocortins. J. Biol. Chem. 285, 17271–17276 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. do Carmo, J. M. et al. Control of metabolic and cardiovascular function by the leptin-brain melanocortin pathway. IUBMB Life 65, 692–698 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Snitker, S., Pratley, R. E., Nicolson, M., Tataranni, P. A. & Ravussin, E. Relationship between muscle sympathetic nerve activity and plasma leptin concentration. Obes. Res. 5, 338–340 (1997).

    Article  CAS  PubMed  Google Scholar 

  124. Haynes, W. G., Morgan, D. A., Walsh, S. A., Mark, A. L. & Sivitz, W. I. Receptor-mediated regional sympathetic nerve activation by leptin. J. Clin. Invest. 100, 270–278 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Shek, E. W., Brands, M. W. & Hall, J. E. Chronic leptin infusion increases arterial pressure. Hypertension 31, 409–414 (1998).

    Article  CAS  PubMed  Google Scholar 

  126. Mark, A. L. Selective leptin resistance revisited. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305, R566–R581 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Kuo, J. J., Jones, O. B. & Hall, J. E. Inhibition of NO synthesis enhances chronic cardiovascular and renal actions of leptin. Hypertension 37, 670–676 (2001).

    Article  CAS  PubMed  Google Scholar 

  128. Carlyle, M., Jones, O. B., Kuo, J. J. & Hall, J. E. Chronic cardiovascular and renal actions of leptin: role of adrenergic activity. Hypertension 39, 496–501 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Lim, K., Burke, S. L. & Head, G. A. Obesity-related hypertension and the role of insulin and leptin in high-fat-fed rabbits. Hypertension 61, 628–634 (2013).

    Article  CAS  PubMed  Google Scholar 

  130. Mark, A. L. et al. Contrasting blood pressure effects of obesity in leptin-deficient ob/ob mice and agouti yellow obese mice. J. Hypertens. 17, 1949–1953 (1999).

    Article  CAS  PubMed  Google Scholar 

  131. Aizawa-Abe, M. et al. Pathophysiological role of leptin in obesity-related hypertension. J. Clin. Invest. 105, 1243–1252 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Machleidt, F. et al. Experimental hyperleptinemia acutely increases vasoconstrictory sympathetic nerve activity in healthy humans. J. Clin. Endocrinol. Metab. 98, E491–E496 (2013).

    Article  CAS  PubMed  Google Scholar 

  133. Zelissen, P. M. et al. Effect of three treatment schedules of recombinant methionyl human leptin on body weight in obese adults: a randomized, placebo-controlled trial. Diabetes Obes. Metab. 7, 755–761 (2005).

    Article  CAS  PubMed  Google Scholar 

  134. Ozata, M., Ozdemir, I. C. & Licinio, J. Human leptin deficiency caused by a missense mutation: multiple endocrine defects, decreased sympathetic tone, and immune system dysfunction indicate new targets for leptin action, greater central than peripheral resistance to the effects of leptin, and spontaneous correction of leptin-mediated defects. J. Clin. Endocrinol. Metab. 84, 3686–3695 (1999).

    Article  CAS  PubMed  Google Scholar 

  135. Munzberg, H. & Myers, M. G. Jr. Molecular and anatomical determinants of central leptin resistance. Nat. Neurosci. 8, 566–570 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Rahmouni, K., Morgan, D. A., Morgan, G. M., Mark, A. L. & Haynes, W. G. Role of selective leptin resistance in diet-induced obesity hypertension. Diabetes 54, 2012–2018 (2005).

    Article  CAS  PubMed  Google Scholar 

  137. Dubinion, J. H., da Silva, A. A. & Hall, J. E. Chronic blood pressure and appetite responses to central leptin infusion in rats fed a high fat diet. J. Hypertens. 29, 758–762 (2011).

    Article  CAS  PubMed  Google Scholar 

  138. Mark, A. L. et al. Leptin signaling in the nucleus tractus solitarii increases sympathetic nerve activity to the kidney. Hypertension 53, 375–380 (2009).

    Article  CAS  PubMed  Google Scholar 

  139. Harlan, S. M. et al. Ablation of the leptin receptor in the hypothalamic arcuate nucleus abrogates leptin-induced sympathetic activation. Circ. Res. 108, 808–812 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. do Carmo, J. M. et al. Control of blood pressure, appetite, and glucose by leptin in mice lacking leptin receptors in proopiomelanocortin neurons. Hypertension 57, 918–926 (2011).

    Article  CAS  PubMed  Google Scholar 

  141. Gao, Q. et al. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc. Natl Acad. Sci. USA 101, 4661–4666 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Dubinion, J. H. et al. Role of proopiomelanocortin neuron Stat3 in regulating arterial pressure and mediating the chronic effects of leptin. Hypertension 61, 1066–1074 (2013).

    Article  CAS  PubMed  Google Scholar 

  143. Rahmouni, K., Haynes, W. G., Morgan, D. A. & Mark, A. L. Intracellular mechanisms involved in leptin regulation of sympathetic outflow. Hypertension 41, 763–767 (2003).

    Article  CAS  PubMed  Google Scholar 

  144. do Carmo, J. M., da Silva, A. A. & Hall, J. E. Leptin reduces food intake but fails to raise blood pressure in mice with deficiency of insulin receptor substrate 2 (IRS2) in the entire brain or specifically in POMC neurons. Hypertension 60, A27 (2012).

    Google Scholar 

  145. Krajewska, M. et al. Development of diabesity in mice with neuronal deletion of Shp2 tyrosine phosphatase. Am. J. Pathol. 172, 1312–1324 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. do Carmo, J. M. et al. Shp2 signaling in POMC neurons is important for leptin’s actions on blood pressure, energy balance, and glucose regulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 307, R1438–R1447 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. do Carmo, J. M. et al. Role of Shp2 in forebrain neurons in regulating metabolic and cardiovascular functions and responses to leptin. Int. J. Obes. 38, 775–783 (2014).

    Article  CAS  Google Scholar 

  148. Belin de Chantemele, E. J. et al. Protein tyrosine phosphatase 1B, a major regulator of leptin-mediated control of cardiovascular function. Circulation 120, 753–763 (2009).

    Article  CAS  PubMed  Google Scholar 

  149. Aberdein, N. et al. Role of PTP1B in POMC neurons during chronic high-fat diet: sex differences in regulation of liver lipids and glucose tolerance. Am. J. Physiol. Regul. Integr. Comp. Physiol. 314, R478–R488 (2018).

    Article  CAS  PubMed  Google Scholar 

  150. Mori, H. et al. Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat. Med. 10, 739–743 (2004).

    Article  CAS  PubMed  Google Scholar 

  151. do Carmo, J. M. et al. Neuronal suppressor of cytokine signaling 3: role in modulating chronic metabolic and cardiovascular effects of leptin. Hypertension 71, 1248–1257 (2018).

    Article  CAS  PubMed  Google Scholar 

  152. Grassi, G., Mark, A. & Esler, M. The sympathetic nervous system alterations in human hypertension. Circ. Res. 116, 976–990 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Gentile, C. L., Orr, J. S., Davy, B. M. & Davy, K. P. Modest weight gain is associated with sympathetic neural activation in nonobese humans. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292, R1834–R1838 (2007).

    Article  CAS  PubMed  Google Scholar 

  154. Esler, M. et al. Obesity paradox in hypertension: is this because sympathetic activation in obesity-hypertension takes a benign form? Hypertension 71, 22–33 (2018).

    Article  CAS  PubMed  Google Scholar 

  155. Armitage, J. A. et al. Rapid onset of renal sympathetic nerve activation in rabbits fed a high-fat diet. Hypertension 60, 163–171 (2012).

    Article  CAS  PubMed  Google Scholar 

  156. Davy, K. P. & Hall, J. E. Obesity and hypertension: two epidemics or one? Am. J. Physiol. Regul. Integr. Comp. Physiol. 286, R803–R813 (2004).

    Article  CAS  PubMed  Google Scholar 

  157. Davy, K. P. & Orr, J. S. Sympathetic nervous system behavior in human obesity. Neurosci. Biobehav. Rev. 33, 116–124 (2009).

    Article  PubMed  Google Scholar 

  158. Wofford, M. R. et al. Antihypertensive effect of alpha- and beta-adrenergic blockade in obese and lean hypertensive subjects. Am. J. Hypertens. 14, 694–698 (2001).

    Article  CAS  PubMed  Google Scholar 

  159. Kassab, S. et al. Endothelin-A receptor antagonism attenuates the hypertension and renal injury in Dahl salt-sensitive rats. Hypertension 31, 397–402 (1998).

    Article  CAS  PubMed  Google Scholar 

  160. Lohmeier, T. E. et al. Systemic and renal-specific sympathoinhibition in obesity hypertension. Hypertension 59, 331–338 (2012).

    Article  CAS  PubMed  Google Scholar 

  161. Henegar, J. R. et al. Catheter-based radiorefrequency renal denervation lowers blood pressure in obese hypertensive dogs. Am. J. Hypertens. 27, 1285–1292 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Iliescu, R., Lohmeier, T. E., Tudorancea, I., Laffin, L. & Bakris, G. L. Renal denervation for the treatment of resistant hypertension: review and clinical perspective. Am. J. Physiol. Renal Physiol. 309, F583–F594 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Asirvatham-Jeyaraj, N. et al. Renal denervation normalizes arterial pressure with no effect on glucose metabolism or renal inflammation in obese hypertensive mice. Hypertension 68, 929–936 (2016).

    Article  CAS  PubMed  Google Scholar 

  164. Hering, D. et al. Sustained sympathetic and blood pressure reduction 1 year after renal denervation in patients with resistant hypertension. Hypertension 64, 118–124 (2014).

    Article  CAS  PubMed  Google Scholar 

  165. Bhatt, D. L. et al. A controlled trial of renal denervation for resistant hypertension. N. Engl. J. Med. 370, 1393–1401 (2014).

    Article  CAS  PubMed  Google Scholar 

  166. Azizi, M. et al. Endovascular ultrasound renal denervation to treat hypertension (RADIANCE-HTN SOLO): a multicentre, international, single-blind, randomised, sham-controlled trial. Lancet 391, 2335–2345 (2018).

    Article  PubMed  Google Scholar 

  167. Henegar, J. R. et al. Catheter-based radiofrequency renal denervation: location effects on renal norepinephrine. Am. J. Hypertens. 28, 909–914 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Kandzari, D. E. et al. Effect of renal denervation on blood pressure in the presence of antihypertensive drugs: 6-month efficacy and safety results from the SPYRAL HTN-ON MED proof-of-concept randomised trial. Lancet 391, 2346–2355 (2018).

    Article  PubMed  Google Scholar 

  169. DiBona, G. F. Sympathetic nervous system and hypertension. Hypertension 61, 556–560 (2013).

    Article  CAS  PubMed  Google Scholar 

  170. Schlaich, M. P., Sobotka, P. A., Krum, H., Lambert, E. & Esler, M. D. Renal sympathetic-nerve ablation for uncontrolled hypertension. N. Engl. J. Med. 361, 932–934 (2009).

    Article  CAS  PubMed  Google Scholar 

  171. Hering, D. et al. Substantial reduction in single sympathetic nerve firing after renal denervation in patients with resistant hypertension. Hypertension 61, 457–464 (2013).

    Article  CAS  PubMed  Google Scholar 

  172. Brinkmann, J. et al. Catheter-based renal nerve ablation and centrally generated sympathetic activity in difficult-to-control hypertensive patients: prospective case series. Hypertension 60, 1485–1490 (2012).

    Article  CAS  PubMed  Google Scholar 

  173. Zappe, D. H. et al. Role of renal afferent nerves in obesity-induced hypertension. Am. J. Hypertens. 9, 20A (1996).

    Article  Google Scholar 

  174. Dewan, N. A., Nieto, F. J. & Somers, V. K. Intermittent hypoxemia and OSA: implications for comorbidities. Chest 147, 266–274 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Lim, K., Jackson, K. L., Sata, Y. & Head, G. A. Factors responsible for obesity-related hypertension. Curr. Hypertens. Rep. 19, 53 (2017).

    Article  PubMed  Google Scholar 

  176. Hall, J. E., Hildebrandt, D. A. & Kuo, J. Obesity hypertension: role of leptin and sympathetic nervous system. Am. J. Hypertens. 14, 103S–115S (2001).

    Article  CAS  PubMed  Google Scholar 

  177. Hall, J. E. Hyperinsulinemia: a link between obesity and hypertension? Kidney Int. 43, 1402–1417 (1993).

    Article  CAS  PubMed  Google Scholar 

  178. Hall, J. E., Brands, M. W., Hildebrandt, D. A. & Mizelle, H. L. Obesity-associated hypertension. Hyperinsulinemia and renal mechanisms. Hypertension 19, I45–I55 (1992).

    CAS  PubMed  Google Scholar 

  179. Iliescu, R., Tudorancea, I., Irwin, E. D. & Lohmeier, T. E. Chronic baroreflex activation restores spontaneous baroreflex control and variability of heart rate in obesity-induced hypertension. Am. J. Physiol. Heart Circ. Physiol. 305, H1080–H1088 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Seravalle, G. et al. Long-term sympathoinhibitory effects of surgically induced weight loss in severe obese patients. Hypertension 64, 431–437 (2014).

    Article  CAS  PubMed  Google Scholar 

  181. Grassi, G. et al. Body weight reduction, sympathetic nerve traffic, and arterial baroreflex in obese normotensive humans. Circulation 97, 2037–2042 (1998).

    Article  CAS  PubMed  Google Scholar 

  182. Mansukhani, M. P., Kara, T., Caples, S. M. & Somers, V. K. Chemoreflexes, sleep apnea, and sympathetic dysregulation. Curr. Hypertens. Rep. 16, 476 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Mansukhani, M. P., Wang, S. & Somers, V. K. Chemoreflex physiology and implications for sleep apnoea: insights from studies in humans. Exp. Physiol. 100, 130–135 (2015).

    Article  PubMed  Google Scholar 

  184. Floras, J. S. Sleep apnea and cardiovascular disease: an enigmatic risk factor. Circ. Res. 122, 1741–1764 (2018).

    Article  CAS  PubMed  Google Scholar 

  185. Javaheri, S. et al. Sleep apnea: types, mechanisms, and clinical cardiovascular consequences. J. Am. Coll. Cardiol. 69, 841–858 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Iturriaga, R., Oyarce, M. P. & Dias, A. C. R. Role of carotid body in intermittent hypoxia-related hypertension. Curr. Hypertens. Rep. 19, 38 (2017).

    Article  CAS  PubMed  Google Scholar 

  187. Ciriello, J. & Moreau, J. M. Leptin signaling in the nucleus of the solitary tract alters the cardiovascular responses to activation of the chemoreceptor reflex. Am. J. Physiol. Regul. Integr. Comp. Physiol. 303, R727–R736 (2012).

    Article  CAS  PubMed  Google Scholar 

  188. Lohmeier, T. E. et al. Chronic interactions between carotid baroreceptors and chemoreceptors in obesity hypertension. Hypertension 68, 227–235 (2016).

    Article  CAS  PubMed  Google Scholar 

  189. do Carmo, J. M. et al. Role of the brain melanocortins in blood pressure regulation. Biochim. Biophys. Acta 1863, 2508–2514 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  190. da Silva, A. A., do Carmo, J. M., Wang, Z. & Hall, J. E. The brain melanocortin system, sympathetic control, and obesity hypertension. Physiology 29, 196–202 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Tao, Y. X. The melanocortin-4 receptor: physiology, pharmacology, and pathophysiology. Endocr. Rev. 31, 506–543 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Cone, R. D. Studies on the physiological functions of the melanocortin system. Endocr. Rev. 27, 736–749 (2006).

    Article  CAS  PubMed  Google Scholar 

  193. Girardet, C. & Butler, A. A. Neural melanocortin receptors in obesity and related metabolic disorders. Biochim. Biophys. Acta 1842, 482–494 (2014).

    Article  CAS  PubMed  Google Scholar 

  194. Vaisse, C. et al. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J. Clin. Invest. 106, 253–262 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Kuo, J. J., Silva, A. A. & Hall, J. E. Hypothalamic melanocortin receptors and chronic regulation of arterial pressure and renal function. Hypertension 41, 768–774 (2003).

    Article  CAS  PubMed  Google Scholar 

  196. Kuo, J. J., da Silva, A. A., Tallam, L. S. & Hall, J. E. Role of adrenergic activity in pressor responses to chronic melanocortin receptor activation. Hypertension 43, 370–375 (2004).

    Article  CAS  PubMed  Google Scholar 

  197. da Silva, A. A. et al. Endogenous melanocortin system activity contributes to the elevated arterial pressure in spontaneously hypertensive rats. Hypertension 51, 884–890 (2008).

    Article  CAS  PubMed  Google Scholar 

  198. Tallam, L. S., da Silva, A. A. & Hall, J. E. Melanocortin-4 receptor mediates chronic cardiovascular and metabolic actions of leptin. Hypertension 48, 58–64 (2006).

    Article  CAS  PubMed  Google Scholar 

  199. Greenfield, J. R. et al. Modulation of blood pressure by central melanocortinergic pathways. N. Engl. J. Med. 360, 44–52 (2009).

    Article  CAS  PubMed  Google Scholar 

  200. Greenfield, J. R. Melanocortin signalling and the regulation of blood pressure in human obesity. J. Neuroendocrinol. 23, 186–193 (2011).

    Article  CAS  PubMed  Google Scholar 

  201. do Carmo, J. M., da Silva, A. A., Rushing, J. S. & Hall, J. E. Activation of the central melanocortin system contributes to the increased arterial pressure in obese Zucker rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 302, R561–R567 (2012).

    Article  CAS  PubMed  Google Scholar 

  202. Ward, K. R., Bardgett, J. F., Wolfgang, L. & Stocker, S. D. Sympathetic response to insulin is mediated by melanocortin 3/4 receptors in the hypothalamic paraventricular nucleus. Hypertension 57, 435–441 (2011).

    Article  CAS  PubMed  Google Scholar 

  203. Yosten, G. L., Pate, A. T. & Samson, W. K. Neuronostatin acts in brain to biphasically increase mean arterial pressure through sympatho-activation followed by vasopressin secretion: the role of melanocortin receptors. Am. J. Physiol. Regul. Integr. Comp. Physiol. 300, R1194–R1199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Yosten, G. L. & Samson, W. K. Neural circuitry underlying the central hypertensive action of nesfatin-1: melanocortins, corticotropin-releasing hormone, and oxytocin. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306, R722–R727 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. da Silva, A. A. et al. Chronic central nervous system MC3/4R blockade attenuates hypertension induced by nitric oxide synthase inhibition but not by angiotensin II infusion. Hypertension 65, 171–177 (2014).

    Article  CAS  PubMed  Google Scholar 

  206. Sayk, F. et al. Sympathetic function in human carriers of melanocortin-4 receptor gene mutations. J. Clin. Endocrinol. Metab. 95, 1998–2002 (2010).

    Article  CAS  PubMed  Google Scholar 

  207. Rossi, J. et al. Melanocortin-4 receptors expressed by cholinergic neurons regulate energy balance and glucose homeostasis. Cell Metab. 13, 195–204 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Sohn, J. W. et al. Melanocortin 4 receptors reciprocally regulate sympathetic and parasympathetic preganglionic neurons. Cell 152, 612–619 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. do Carmo, J. M., da Silva, A. A., Rushing, J. S., Pace, B. & Hall, J. E. Differential control of metabolic and cardiovascular functions by melanocortin-4 receptors in proopiomelanocortin neurons. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305, R359–R368 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Nicholson, J. R., Peter, J. C., Lecourt, A. C., Barde, Y. A. & Hofbauer, K. G. Melanocortin-4 receptor activation stimulates hypothalamic brain-derived neurotrophic factor release to regulate food intake, body temperature and cardiovascular function. J. Neuroendocrinol. 19, 974–982 (2007).

    Article  CAS  PubMed  Google Scholar 

  211. Bariohay, B. et al. Brain-derived neurotrophic factor/tropomyosin-related kinase receptor type B signaling is a downstream effector of the brainstem melanocortin system in food intake control. Endocrinology 150, 2646–2653 (2009).

    Article  CAS  PubMed  Google Scholar 

  212. Hall, J. E., Brands, M. W., Dixon, W. N. & Smith, M. J. Jr. Obesity-induced hypertension. Renal function and systemic hemodynamics. Hypertension 22, 292–299 (1993).

    CAS  PubMed  Google Scholar 

  213. Carroll, J. F., Huang, M., Hester, R. L., Cockrell, K. & Mizelle, H. L. Hemodynamic alterations in hypertensive obese rabbits. Hypertension 26, 465–470 (1995).

    Article  CAS  PubMed  Google Scholar 

  214. da Silva, A. A., Kuo, J. J., Tallam, L. S. & Hall, J. E. Role of endothelin-1 in blood pressure regulation in a rat model of visceral obesity and hypertension. Hypertension 43, 383–387 (2004).

    Article  CAS  PubMed  Google Scholar 

  215. Mills, E., Kuhn, C. M., Feinglos, M. N. & Surwit, R. Hypertension in CB57BL/6J mouse model of non-insulin-dependent diabetes mellitus. Am. J. Physiol. 264, R73–R78 (1993).

    CAS  PubMed  Google Scholar 

  216. Aronne, L. J., Mackintosh, R., Rosenbaum, M., Leibel, R. L. & Hirsch, J. Autonomic nervous system activity in weight gain and weight loss. Am. J. Physiol. 269, R222–R225 (1995).

    CAS  Google Scholar 

  217. Contreras, R. J. & Williams, V. L. Dietary obesity and weight cycling: effects on blood pressure and heart rate in rats. Am. J. Physiol. 256, R1209–R1219 (1989).

    CAS  PubMed  Google Scholar 

  218. Wang, Z. et al. Role of Socs3 in Pomc neurons in metabolic and cardiovascular regulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. https://doi.org/10.1152/ajpregu.00163.2018 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  219. Messerli, F. H. et al. Obesity and essential hypertension. Hemodynamics, intravascular volume, sodium excretion, and plasma renin activity. Arch. Intern. Med. 141, 81–85 (1981).

    Article  CAS  PubMed  Google Scholar 

  220. Crandall, D. L., Goldstein, B. M., Lizzo, F. H., Gabel, R. A. & Cervoni, P. Hemodynamics of obesity: influence of pattern of adipose tissue cellularity. Am. J. Physiol. 251, R314–R319 (1986).

    CAS  PubMed  Google Scholar 

  221. Adolphe, J. L., Silver, T. I., Childs, H., Drew, M. D. & Weber, L. P. Short-term obesity results in detrimental metabolic and cardiovascular changes that may not be reversed with weight loss in an obese dog model. Br. J. Nutr. 112, 647–656 (2014).

    Article  CAS  PubMed  Google Scholar 

  222. Philip-Couderc, P. et al. Cardiac transcriptome analysis in obesity-related hypertension. Hypertension 41, 414–421 (2003).

    Article  CAS  PubMed  Google Scholar 

  223. Carroll, J. F., Braden, D. S., Cockrell, K. & Mizelle, H. L. Obese hypertensive rabbits develop concentric and eccentric hypertrophy and diastolic filling abnormalities. Am. J. Hypertens. 10, 230–233 (1997).

    Article  CAS  PubMed  Google Scholar 

  224. Nagarajan, V. et al. Cardiac function and lipid distribution in rats fed a high-fat diet: in vivo magnetic resonance imaging and spectroscopy. Am. J. Physiol. Heart Circ. Physiol. 304, H1495–H1504 (2013).

    Article  CAS  PubMed  Google Scholar 

  225. Mendes-Junior, L. G. et al. The usefulness of short-term high-fat/high salt diet as a model of metabolic syndrome in mice. Life Sci. 209, 341–348 (2018).

    Article  CAS  PubMed  Google Scholar 

  226. Carroll, J. F. et al. Hypertension, cardiac hypertrophy, and neurohumoral activity in a new animal model of obesity. Am. J. Physiol. 271, H373–H378 (1996).

    CAS  PubMed  Google Scholar 

  227. Koncsos, G. et al. Diastolic dysfunction in prediabetic male rats: role of mitochondrial oxidative stress. Am. J. Physiol. Heart Circ. Physiol. 311, H927–H943 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  228. Rocchini, A. P., Moorehead, C., Katch, V., Key, J. & Finta, K. M. Forearm resistance vessel abnormalities and insulin resistance in obese adolescents. Hypertension 19, 615–620 (1992).

    Article  CAS  PubMed  Google Scholar 

  229. Franco, R. L. et al. Forearm blood flow response to acute exercise in obese and non-obese males. Eur. J. Appl. Physiol. 113, 2015–2023 (2013).

    Article  PubMed  Google Scholar 

  230. Chagnac, A. et al. Obesity-induced glomerular hyperfiltration: its involvement in the pathogenesis of tubular sodium reabsorption. Nephrol. Dial. Transplant. 23, 3946–3952 (2008).

    Article  CAS  PubMed  Google Scholar 

  231. D’Agati, V. D. et al. Obesity-related glomerulopathy: clinical and pathologic characteristics and pathogenesis. Nat. Rev. Nephrol. 12, 453–471 (2016).

    Article  CAS  PubMed  Google Scholar 

  232. Antic, V., Tempini, A. & Montani, J. P. Serial changes in cardiovascular and renal function of rabbits ingesting a high-fat, high-calorie diet. Am. J. Hypertens. 12, 826–829 (1999).

    Article  CAS  PubMed  Google Scholar 

  233. Polichnowski, A. J. et al. Glomerulosclerosis in the diet-induced obesity model correlates with sensitivity to nitric oxide inhibition but not glomerular hyperfiltration or hypertrophy. Am. J. Physiol. Renal Physiol. 309, F791–F799 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Riazi, S., Tiwari, S., Sharma, N., Rash, A. & Ecelbarger, C. M. Abundance of the Na-K-2Cl cotransporter NKCC2 is increased by high-fat feeding in Fischer 344 X Brown Norway (F1) rats. Am. J. Physiol. Renal Physiol. 296, F762–F770 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Nizar, J. M. et al. Na+-sensitive elevation in blood pressure is ENaC independent in diet-induced obesity and insulin resistance. Am. J. Physiol. Renal Physiol. 310, F812–F820 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Dwyer, T. M., Bigler, S. A., Moore, N. A., Carroll, J. F. & Hall, J. E. The altered structure of renal papillary outflow tracts in obesity. Ultrastruct. Pathol. 24, 251–257 (2000).

    Article  CAS  PubMed  Google Scholar 

  237. Hall, J. E. et al. Hemodynamic and renal responses to chronic hyperinsulinemia in obese, insulin-resistant dogs. Hypertension 25, 994–1002 (1995).

    Article  CAS  PubMed  Google Scholar 

  238. Eppel, G. A., Armitage, J. A., Eikelis, N., Head, G. A. & Evans, R. G. Progression of cardiovascular and endocrine dysfunction in a rabbit model of obesity. Hypertens. Res. 36, 588–595 (2013).

    Article  CAS  PubMed  Google Scholar 

  239. do Carmo, J. M. et al. Inhibition of soluble epoxide hydrolase reduces food intake and increases metabolic rate in obese mice. Nutr. Metab. Cardiovasc. Dis. 22, 598–604 (2012).

    Article  CAS  PubMed  Google Scholar 

  240. Carroll, J. F. & Kyser, C. K. Exercise training in obesity lowers blood pressure independent of weight change. Med. Sci. Sports Exerc. 34, 596–601 (2002).

    PubMed  Google Scholar 

  241. Northcott, C. A. et al. The development of hypertension and hyperaldosteronism in a rodent model of life-long obesity. Endocrinology 153, 1764–1773 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Li, C. et al. High-fat diet amplifies renal renin angiotensin system expression, blood pressure elevation, and renal dysfunction caused by Ceacam1 null deletion. Am. J. Physiol. Endocrinol. Metab. 309, E802–E810 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Schafer, N. et al. Endothelial mineralocorticoid receptor activation mediates endothelial dysfunction in diet-induced obesity. Eur. Heart J. 34, 3515–3524 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Khan, S. A. et al. Obesity depresses baroreflex control of renal sympathetic nerve activity and heart rate in Sprague Dawley rats: role of the renal innervation. Acta Physiol. 214, 390–401 (2015).

    Article  CAS  Google Scholar 

  245. Fardin, N. M., Oyama, L. M. & Campos, R. R. Changes in baroreflex control of renal sympathetic nerve activity in high-fat-fed rats as a predictor of hypertension. Obesity 20, 1591–1597 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors’ research was supported by grants from the US National Heart, Lung, and Blood Institute (P01 HL51971), the US National Institute of General Medical Sciences (P20 GM104357 and U54 GM115428) and the US National Institute of Diabetes and Digestive and Kidney Diseases (1K08DK099415-01A1) of the US National Institutes of Health.

Reviewer information

Nature Reviews Nephrology thanks the anonymous reviewers for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

J.E.H. wrote the manuscript. All authors researched the data for the article, contributed to discussions of the content and reviewed or edited the manuscript before submission.

Corresponding author

Correspondence to John E. Hall.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Lipotoxicity

The toxic effect of lipids that accumulate in non-adipose tissue and cause cellular dysfunction.

Renal sinus fat

(RSF). The adipose tissue that accumulates in the renal sinuses, which are cavities within the kidneys that are occupied by the renal pelvis, renal calyces, blood vessels and nerves.

Perirenal fat

(PRF). Also called the adipose capsule of the kidney. The perirenal fat is a structure located between the renal fascia and renal capsule.

Blood flow reserve

The maximum increase in blood flow above the resting level of blood flow.

Hypoxaemia

The condition of abnormally low oxygen concentration in the blood.

Baroreflex

Often called the baroreceptor reflex. The reflex mechanism by which stretch receptors (baroreceptors), located especially in the carotid sinuses and aortic arch, regulate blood pressure.

Hypercapnia

The condition of excessive carbon dioxide concentration in the blood.

Eucapnic

The condition of having normal carbon dioxide concentration in the blood.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hall, J.E., do Carmo, J.M., da Silva, A.A. et al. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 15, 367–385 (2019). https://doi.org/10.1038/s41581-019-0145-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41581-019-0145-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing