Sex Disparity in Cardiovascular Disease: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Donato Gemmati.

Among cardiovascular diseases, acute myocardial infarction (AMI) shows significant differences in occurrence rate, prognosis and efficacy of treatment between male and female patients. Genomics and epigenomics approaches together with epidrugs design and drug repositioning could fill the sex-gap.

  • gender medicine
  • sex disparities
  • genetics/molecular biomarkers
  • molecular medicine
  • OMICS
  • genomics

1. Introduction

In the recent past, there has been a growing attention to sex-based differences in biology, genetics, biomedical sciences and general medicine, ranging from the cellular level to whole organs and organisms. As expected, this process quickly led to the generation of new insights into diagnostic, prognostic and therapeutic issues, from basic research to the clinical level [17][1]. The overall message is indeed the one published by Nature in 2010, which summarizes old and new problems in the title “Putting gender on the agenda” [18][2]. Starting from the fact that animals have a sex [19][3], well known differences in gene expression have to exist in male versus female mice [20][4]. Based on the evidence that companies and scientists may have arbitrarily performed their preclinical tests on male models, the Editors of Nature concluded that “Medicine as it is currently applied to women is less evidence-based than that being applied to men” [18][2]. The increasing attention towards sex and gender, along with the interest that emerges from this kind of aware research, are now beginning to bridge the gap [21][5]. Thanks to the increased knowledge of the molecular, genomics and epigenomics bases of complex diseases, and thanks to the personalized pharmacogenetic/genomics approach to drug design/prescription, several diseases are now faced in a tailored fashion [22,23][6][7]. However, while the inclusion of sex is a process already underway, with evident results from both preclinical and clinical trials, the impact of gender in medical/biomedical fields is still at an early stage, with difficulties and delays due to its intrinsic complexity. Ongoing efforts aim to include and understand the role of gender in pharmacology [24,25][8][9]. To date, gender-related pharmacodynamic and pharmacokinetic differences have been reported with crucial implications on drugs effects [26,27,28,29,30][10][11][12][13][14]. Overall, gender-specific pre-clinical models will increase the definition of gender-oriented therapeutic protocols, in turn accelerating the development of gender-specific drugs and the generation of gender-oriented and evidence-based guidelines [11,31][15][16].

2. Sex Disparity

Improvement in the acute treatment has led to a dramatic increasing in the number of acute myocardial infarction (AMI)survivors among those with damaged heart. These patients are at risk of developing severe complications, like left ventricular remodelling (LVR) and heart failure (HF), which are still considered orphans of specific prognostic tools and effective dedicated treatments [45,46][17][18]. AMI is the most frequent cause of LVR and HF and patients often undergo to chronic and costly therapies, very frequent re-hospitalization and poor quality of life, with a significant gap among the two sexes [9,45,46][19][17][18]. Recently it has been reported that “the incidence and prevalence of coronary artery disease in women has exceeded that in men over the past four decades” [47,48][20][21].
AMI takes place from a complex interrelationship among genetic/epigenetic and environmental risk factors, as it was revealed by several studies of pharmacogenetics and -genomics [49[22][23][24],50,51], and more extensively confirmed by omics investigations [32,33][25][26]. In addition, a very recent study from the Multi-Analyte, Genetic, and Thrombogenic Markers of Atherosclerosis group (MAGMA) found that females with angina are more thrombogenic than males, and this difference may affect sex-related outcomes [52][27], also on the basis of undeniable genomics differences. Moreover, the Genetics of Subsequent Coronary Heart Disease consortium (GENIUS-CHD) was established to discover and validate genetic variants and biomarkers for the risk assessment of subsequent ischemic events along with novel drug targets for secondary prevention. It is considered a first-class initiative able to generate extraordinary results also in the field of sex-oriented cardiology [53][28]. Our group has contributed for a long time to the disclosure of the genetics/pharmacogenetics bases of myocardial infarction [54,55,56[29][30][31][32][33][34],57,58,59], and very recently we summarized our previous efforts and patents [US2016363592 (A1); ITTO20130532 (A1)] in a Special Issue belonging to the “Novel Molecular Targets for Cardioprotection: The EU-Cardioprotection Cost Action (CA16225)”, which suggest useful sex-oriented prognostic biomarkers [60][35].
Noteworthy, the incidence of AMI is much lower among females under the age of 50 years compared with males, but after the menopause, the rate in females dramatically increases, approaching that of males [9,61][19][36]. For this reason, oestrogens were postulated to be cardioprotective but results coming from recent randomized clinical trials challenge this hypothesis [61][36]. In addition, sex differences influence AMI pathophysiology, clinical presentation and clinical outcome. In detail, the mortality rate after one or five years is higher in females than in males, and the former more frequently experience heart wall remodelling, failure and stroke within five years from the first ischemic accident. This remains the poorest outcome, even considering several concomitant situations, like different age at presentation, genetic risk factors and comorbidities, as demonstrated by the higher in-hospital mortality, the readmission rate within the first thirty days or the longer follow-up [9][19]. Interestingly, results coming from five Italian prospective registries conducted between 2001 and 2014, concluded that age and sex specific differences exist in the outcome of patients with ST-elevation myocardial infarction (STEMI) [61,62][36][37]. Regardless of age, at discharge, females are at higher risk of re-hospitalization due to different causes. Strong disparities between the two sexes were confirmed also after adjustment for confounding factors [63][38]. Accordingly, the in-hospital death was 3.2% for males and 8.4% for females, and the latter have been found significantly associated with in-hospital mortality. Finally, females continue to experience higher post-AMI mortality and global poorer outcome (Figure 21) also despite the improvements in reperfusion therapy tools [64][39]. In conclusion, despite a worldwide improvement in STEMI care, females continue to experience higher in-hospital mortality, which is not completely unexpected, as the most fundamental cardiovascular studies are primarily based on males [9,65][19][40].
Figure 21. Extreme clinical phenotypes and prognosis in male and female AMI patients. The increased AMI risk in males is balanced by a better prognosis, resulting in enhanced AMI outcome. Conversely, the reduced AMI risk in females is characterized by a worst prognosis, resulting in a poor AMI outcome.
Another key element is that postmenopausal females tend to develop HF with preserved ejection fraction, characterized by diastolic dysfunction, whereas age-matching males develop HF with reduced ejection fraction, characterized by systolic dysfunction [66][41]. These differences are important because most drugs used to treat HF have been developed to treat those with reduced ejection fraction, and there are few effective treatment options for HF with preserved ejection fraction [67][42]. It is also possible that the aging process affects the heart of males and females differently, so that the latter are predisposed to problems of myocardial relaxation, while males mainly develop pump failure. As a result, males and females show a completely opposite trend in terms of hospitalization index for HF, which increases significantly with the age of the females compared with age-matching males [68][43]. A recent long-term study on LVR after the first AMI reports that, of the total number of patients who experienced LVR after one-year follow-up (31–38% in the two cohorts of the study), females were significantly over-represented (43–46.5% vs 27–36.5% for females and males respectively in the two studies) [69][44]. These data are extremely important since both cohorts of patients in that study had a very high rate of secondary prevention medications after one year (i.e., beta-blockers 90–95% and ACE inhibitors/angiotensin II R-blockers 93–97% respectively in the two studies). Interestingly, sex hormones are associated differently with the morphology of the remodelled heart wall, and a more androgenic profile is responsible for a greater imbalance in the heart mass/volume ratio in females than in males [70][45].
In summary, remodelling is the main determinant of survival after recovery from AMI. Observational clinical and post-mortem/experimental studies suggest important differences between females and males in the cardiac remodelling observed in response to different types of cardiac injuries including infarction. Recommended therapies for AMI in females are similar to those in males, but studies indicate that females are undertreated, which lead to worse outcomes [9,47][19][20]. Interestingly, during the first few hours after AMI, complex local healing processes and inflammatory reactions are crucial in determining the risk of wall remodelling. Soluble molecules, resident and circulating cells and micro-RNAs finely mediate inflammation and regenerative progression by determining the fate of the heart after the infarction [71,72,73,74][46][47][48][49]. Basically, imbalance in any healing process and unrestrained ECM proteolysis cause a delayed remedy, while lasting chronic lesions in any district may affect the normal organ/tissue functions with consequent failure and malfunction [75,76,77][50][51][52]. Since these complex processes are in part genetically determined, molecular regenerative medicine, by providing molecular predictive indicators, might help clinicians to recognize at risk populations also in a sex- or gender-oriented direction [78,79,80,81][53][54][55][56]. Reperfusion therapy and systematic use of evidence-based medications have effects on LVR or HF but they cannot be considered dedicated therapeutic approaches. Prognostic biomarkers able to score cases at higher risk to develop severe post-infarction complications (i.e., females) will allow the early identification of patients for whom the available standard care is not adequate. It is to be taken into account that groups of males at higher risk could also benefit from personalized approaches.
Overall, there is a mandatory need for a “holistic” sex specific approach (i.e., sex-oriented omics-investigations “sex/genderomics”), from the use of in vitro and in vivo preclinical models up to appropriate clinical studies with well-balanced male/female ratio cohorts. This will allow the identification of prognostic biomarkers to effectively counteract sex-disparities and help in designing sex-dedicated therapies [82][57]. If appropriately addressed, these approaches will represent a significant improvement in the cardiovascular disease management, and the potential of sex-specific risk stratification tools will positively influence the worldwide National Health Care Systems.
 

References

  1. Legato, M.J.; Johnson, P.A.; Manson, J.E. Consideration of Sex Differences in Medicine to Improve Health Care and Patient Outcomes. JAMA 2016, 316, 1865–1866.
  2. Putting gender on the agenda. Nature 2010, 465, 665.
  3. Blaustein, J.D. Animals have a sex, and so should titles and methods sections of articles in Endocrinology. Endocrinology 2012, 153, 2539–2540.
  4. Yang, X.; Schadt, E.E.; Wang, S.; Wang, H.; Arnold, A.P.; Ingram-Drake, L.; Drake, T.A.; Lusis, A.J. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res. 2006, 16, 995–1004.
  5. Oertelt-Prigione, S.; Dalibert, L.; Verdonk, P.; Stutz, E.Z.; Klinge, I. Implementation Strategies for Gender-Sensitive Public Health Practice: A European Workshop. J. Womens Health 2017, 26, 1255–1261.
  6. Kaminsky, Z.; Wang, S.C.; Petronis, A. Complex disease, gender and epigenetics. Ann. Med. 2006, 38, 530–544.
  7. Sioud, M.; Melien, O. Treatment options and individualized medicine. Methods Mol. Biol. 2007, 361, 327–340.
  8. Franconi, F.; Raparelli, V.; Regitz-Zagrosek, V. Sex and gender landscape in pharmacology. Pharmacol. Res. 2017, 123, 93–94.
  9. Rodriquez, M.; Aquino, R.P.; D’Ursi, A.M. Is it time to integrate sex and gender into drug design and development? Future Med. Chem. 2015, 7, 557–559.
  10. Franconi, F.; Campesi, I. Sex and gender influences on pharmacological response: An overview. Expert Rev. Clin. Pharmacol. 2014, 7, 469–485.
  11. Basili, S.; Raparelli, V.; Proietti, M.; Tanzilli, G.; Franconi, F. Impact of sex and gender on the efficacy of antiplatelet therapy: The female perspective. J. Atheroscler. Thromb. 2015, 22, 109–125.
  12. Di Giosia, P.; Passacquale, G.; Petrarca, M.; Giorgini, P.; Marra, A.M.; Ferro, A. Gender differences in cardiovascular prophylaxis: Focus on antiplatelet treatment. Pharmacol. Res. 2017, 119, 36–47.
  13. Pucci, G.; Alcidi, R.; Tap, L.; Battista, F.; Mattace-Raso, F.; Schillaci, G. Sex- and gender-related prevalence, cardiovascular risk and therapeutic approach in metabolic syndrome: A review of the literature. Pharmacol. Res. 2017, 120, 34–42.
  14. Campesi, I.; Franconi, F.; Seghieri, G.; Meloni, M. Sex-gender-related therapeutic approaches for cardiovascular complications associated with diabetes. Pharmacol. Res. 2017, 119, 195–207.
  15. Franconi, F.; Rosano, G.; Campesi, I. Need for gender-specific pre-analytical testing: The dark side of the moon in laboratory testing. Int. J. Cardiol. 2015, 179, 514–535.
  16. Franconi, F.; Campesi, I.; Colombo, D.; Antonini, P. Sex-Gender Variable: Methodological Recommendations for Increasing Scientific Value of Clinical Studies. Cells 2019, 8, 476.
  17. Available online: http://www.worldheartfailure.org/WHFS (accessed on 24 October 2019).
  18. Available online: http://www.medicographia.com/2012/02/the-heart-failure-epidemic (accessed on 24 October 2019).
  19. Mehta, L.S.; Beckie, T.M.; DeVon, H.A.; Grines, C.L.; Krumholz, H.M.; Johnson, M.N.; Lindley, K.J.; Vaccarino, V.; Wang, T.Y.; Watson, K.E.; et al. Acute Myocardial Infarction in Women: A Scientific Statement From the American Heart Association. Circulation 2016, 133, 916–947.
  20. Davis, E.; Gorog, D.A.; Rihal, C.; Prasad, A.; Srinivasan, M. “Mind the gap” acute coronary syndrome in women: A contemporary review of current clinical evidence. Int. J. Cardiol. 2017, 227, 840–849.
  21. Oertelt-Prigione, S. Gender and cardiovascular disease in the workplace—It’s not just about pay gaps. Int. J. Cardiol. 2018, 262, 108–109.
  22. Myocardial Infarction Genetics Consortium; Kathiresan, S.; Voight, B.F.; Purcell, S.; Musunuru, K.; Ardissino, D.; Mannucci, P.M.; Anand, S.; Engert, J.C.; Samani, N.J.; et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat. Genet. 2009, 41, 334–341.
  23. Nakatochi, M.; Ichihara, S.; Yamamoto, K.; Naruse, K.; Yokota, S.; Asano, H.; Matsubara, T.; Yokota, M. Epigenome-wide association of myocardial infarction with DNA methylation sites at loci related to cardiovascular disease. Clin. Epigenet. 2017, 9, 54.
  24. Webb, T.R.; Erdmann, J.; Stirrups, K.E.; Stitziel, N.O.; Masca, N.G.; Jansen, H.; Kanoni, S.; Nelson, C.P.; Ferrario, P.G.; Konig, I.R.; et al. Systematic Evaluation of Pleiotropy Identifies 6 Further Loci Associated With Coronary Artery Disease. J. Am. Coll. Cardiol. 2017, 69, 823–836.
  25. Chen, R.; Snyder, M. Promise of personalized omics to precision medicine. Wiley Interdiscip. Rev. Syst. Biol. Med. 2013, 5, 73–82.
  26. Baetta, R.; Pontremoli, M.; Fernandez, A.M.; Spickett, C.M.; Banfi, C. Reprint of: Proteomics in cardiovascular diseases: Unveiling sex and gender differences in the era of precision medicine. J. Proteom. 2018, 178, 57–72.
  27. Chaudhary, R.; Sukhi, A.; Chaudhary, R.; Jindal, M.; Vyas, A.; Rout, A.; Bliden, K.; Tantry, U.; Gurbel, P. Gender differences in thrombogenicity among patients with angina and non-obstructive coronary artery disease. J. Thromb. Thrombolysis 2019.
  28. Patel, R.; Tragante, V.; Schmidt, A.F.; McCubrey, R.O.; Holmes, M.V.; Howe, L.J.; Direk, K.; Akerblom, A.; Leander, K.; Virani, S.S.; et al. Subsequent Event Risk in Individuals with Established Coronary Heart Disease: Design and Rationale of the GENIUS-CHD Consortium. Circ. Genom. Precis. Med. 2019.
  29. Gemmati, D.; Serino, M.L.; Trivellato, C.; Fiorini, S.; Scapoli, G.L. C677T substitution in the methylenetetrahydrofolate reductase gene as a risk factor for venous thrombosis and arterial disease in selected patients. Haematologica 1999, 84, 824–828.
  30. Gemmati, D.; Serino, M.L.; Ongaro, A.; Tognazzo, S.; Moratelli, S.; Resca, R.; Moretti, M.; Scapoli, G.L. A common mutation in the gene for coagulation factor XIII-A (VAL34Leu): A risk factor for primary intracerebral hemorrhage is protective against atherothrombotic diseases. Am. J. Hematol. 2001, 67, 183–188.
  31. Campo, G.; Valgimigli, M.; Ferraresi, P.; Malagutti, P.; Baroni, M.; Arcozzi, C.; Gemmati, D.; Percoco, G.; Parrinello, G.; Ferrari, R.; et al. Tissue factor and coagulation factor VII levels during acute myocardial infarction: Association with genotype and adverse events. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2800–2806.
  32. Gemmati, D.; Federici, F.; Campo, G.; Tognazzo, S.; Serino, M.L.; De Mattei, M.; Valgimigli, M.; Malagutti, P.; Guardigli, G.; Ferraresi, P.; et al. Factor XIIIA-V34L and factor XIIIB-H95R gene variants: Effects on survival in myocardial infarction patients. Mol. Med. 2007, 13, 112–120.
  33. Gemmati, D.; Zeri, G.; Orioli, E.; Mari, R.; Moratelli, S.; Vigliano, M.; Marchesini, J.; Grossi, M.E.; Pecoraro, A.; Cuneo, A.; et al. Factor XIII-A dynamics in acute myocardial infarction: A novel prognostic biomarker? Thromb. Haemost. 2015, 114, 123–132.
  34. Gemmati, D.; Vigliano, M.; Burini, F.; Mari, R.; El Mohsein, H.H.; Parmeggiani, F.; Serino, M.L. Coagulation Factor XIIIA (F13A1): Novel Perspectives in Treatment and Pharmacogenetics. Curr. Pharm. Des. 2016, 22, 1449–1459.
  35. Ansani, L.; Marchesini, J.; Pestelli, G.; Luisi, G.A.; Scillitani, G.; Longo, G.; Milani, D.; Serino, M.L.; Tisato, V.; Gemmati, D. F13A1 Gene Variant (V34L) and Residual Circulating FXIIIA Levels Predict Short- and Long-Term Mortality in Acute Myocardial Infarction after Coronary Angioplasty. Int. J. Mol. Sci. 2018, 19, 2766.
  36. De Luca, L.; Marini, M.; Gonzini, L.; Boccanelli, A.; Casella, G.; Chiarella, F.; De Servi, S.; Di Chiara, A.; Di Pasquale, G.; Olivari, Z.; et al. Contemporary Trends and Age-Specific Sex Differences in Management and Outcome for Patients With ST-Segment Elevation Myocardial Infarction. J. Am. Heart Assoc. 2016, 5.
  37. Dreyer, R.P.; Ranasinghe, I.; Wang, Y.; Dharmarajan, K.; Murugiah, K.; Nuti, S.V.; Hsieh, A.F.; Spertus, J.A.; Krumholz, H.M. Sex Differences in the Rate, Timing, and Principal Diagnoses of 30-Day Readmissions in Younger Patients with Acute Myocardial Infarction. Circulation 2015, 132, 158–166.
  38. Dreyer, R.P.; Dharmarajan, K.; Kennedy, K.F.; Jones, P.G.; Vaccarino, V.; Murugiah, K.; Nuti, S.V.; Smolderen, K.G.; Buchanan, D.M.; Spertus, J.A.; et al. Sex Differences in 1-Year All-Cause Rehospitalization in Patients After Acute Myocardial Infarction: A Prospective Observational Study. Circulation 2017, 135, 521–531.
  39. Writing Group, M.; Mozaffarian, D.; Benjamin, E.J.; Go, A.S.; Arnett, D.K.; Blaha, M.J.; Cushman, M.; Das, S.R.; de Ferranti, S.; Despres, J.P.; et al. Heart Disease and Stroke Statistics-2016 Update: A Report From the American Heart Association. Circulation 2016, 133, e38–e48.
  40. Wells, G.L. Cardiovascular Risk Factors: Does Sex Matter? Curr. Vasc. Pharmacol. 2016, 14, 452–457.
  41. Dunlay, S.M.; Roger, V.L. Gender differences in the pathophysiology, clinical presentation, and outcomes of ischemic heart failure. Curr. Heart Fail. Rep. 2012, 9, 267–276.
  42. Seeland, U.; Regitz-Zagrosek, V. Sex and gender differences in cardiovascular drug therapy. Handb. Exp. Pharmacol. 2012, 211–236.
  43. Ranasinghe, I.; Wang, Y.; Dharmarajan, K.; Hsieh, A.F.; Bernheim, S.M.; Krumholz, H.M. Readmissions after hospitalization for heart failure, acute myocardial infarction, or pneumonia among young and middle-aged adults: A retrospective observational cohort study. PLoS Med. 2014, 11, e1001737.
  44. Bauters, C.; Dubois, E.; Porouchani, S.; Saloux, E.; Fertin, M.; de Groote, P.; Lamblin, N.; Pinet, F. Long-term prognostic impact of left ventricular remodeling after a first myocardial infarction in modern clinical practice. PLoS ONE 2017, 12, e0188884.
  45. Subramanya, V.; Zhao, D.; Ouyang, P.; Lima, J.A.; Vaidya, D.; Ndumele, C.E.; Bluemke, D.A.; Shah, S.J.; Guallar, E.; Nwabuo, C.C.; et al. Sex hormone levels and change in left ventricular structure among men and post-menopausal women: The Multi-Ethnic Study of Atherosclerosis (MESA). Maturitas 2018, 108, 37–44.
  46. Ter Horst, E.N.; Hakimzadeh, N.; van der Laan, A.M.; Krijnen, P.A.; Niessen, H.W.; Piek, J.J. Modulators of Macrophage Polarization Influence Healing of the Infarcted Myocardium. Int. J. Mol. Sci. 2015, 16, 29583–29591.
  47. Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Cardiac Extracellular Vesicles in Normal and Infarcted Heart. Int. J. Mol. Sci. 2016, 17, 63.
  48. Musial-Wysocka, A.; Kot, M.; Sulkowski, M.; Majka, M. Regenerative Potential of the Product “CardioCell” Derived from the Wharton’s Jelly Mesenchymal Stem Cells for Treating Hindlimb Ischemia. Int. J. Mol. Sci. 2019, 20, 4632.
  49. Sun, T.; Dong, Y.H.; Du, W.; Shi, C.Y.; Wang, K.; Tariq, M.A.; Wang, J.X.; Li, P.F. The Role of MicroRNAs in Myocardial Infarction: From Molecular Mechanism to Clinical Application. Int. J. Mol. Sci. 2017, 18, 745.
  50. Singh, A.V.; Subhashree, L.; Milani, P.; Gemmati, D.; Zamboni, P. Interplay of iron metallobiology, metalloproteinases, and FXIII, and role of their gene variants in venous leg ulcer. Int. J. Low Extrem. Wounds 2010, 9, 166–179.
  51. Zamboni, P.; Gemmati, D. Clinical implications of gene polymorphisms in venous leg ulcer: A model in tissue injury and reparative process. Thromb. Haemost. 2007, 98, 131–137.
  52. Zamboni, P.; De Mattei, M.; Ongaro, A.; Fogato, L.; Carandina, S.; De Palma, M.; Tognazzo, S.; Scapoli, G.L.; Serino, M.L.; Caruso, A.; et al. Factor XIII contrasts the effects of metalloproteinases in human dermal fibroblast cultured cells. Vasc. Endovasc. Surg. 2004, 38, 431–438.
  53. Tognazzo, S.; Gemmati, D.; Palazzo, A.; Catozzi, L.; Carandina, S.; Legnaro, A.; Tacconi, G.; Scapoli, G.L.; Zamboni, P. Prognostic role of factor XIII gene variants in nonhealing venous leg ulcers. J. Vasc. Surg. 2006, 44, 815–819.
  54. Gemmati, D.; Tognazzo, S.; Catozzi, L.; Federici, F.; De Palma, M.; Gianesini, S.; Scapoli, G.L.; De Mattei, M.; Liboni, A.; Zamboni, P. Influence of gene polymorphisms in ulcer healing process after superficial venous surgery. J. Vasc. Surg. 2006, 44, 554–562.
  55. Gemmati, D.; Tognazzo, S.; Serino, M.L.; Fogato, L.; Carandina, S.; De Palma, M.; Izzo, M.; De Mattei, M.; Ongaro, A.; Scapoli, G.L.; et al. Factor XIII V34L polymorphism modulates the risk of chronic venous leg ulcer progression and extension. Wound Repair Regen. 2004, 12, 512–517.
  56. Gemmati, D.; Occhionorelli, S.; Tisato, V.; Vigliano, M.; Longo, G.; Gonelli, A.; Sibilla, M.G.; Serino, M.L.; Zamboni, P. Inherited genetic predispositions in F13A1 and F13B genes predict abdominal adhesion formation: Identification of gender prognostic indicators. Sci. Rep. 2018, 8, 16916.
  57. Greiten, L.E.; Holditch, S.J.; Arunachalam, S.P.; Miller, V.M. Should there be sex-specific criteria for the diagnosis and treatment of heart failure? J. Cardiovasc. Transl. Res. 2014, 7, 139–155.
More
Video Production Service