Anda di halaman 1dari 16

J OU R NA LOFTHE AM ERIC AN COLLEGEOFCARDIOL OG Y VOL. 6 9, NO.

1 5, 2 0 1 7

ª 2 0 1 7 byThe AM ERIC AN CO LLEGEOFCARDIOL OG YF OU N DA TI ON ISSN 0 7 05-01 Maret 0 9 7 / $ 3 6. 0 0

PUBLISHEDBYELSEVIER http: / / dx. doi. org / 1 0. 1 0 1 6 / j. JACC. 2 0 1 7. 0 1. 0 6 4

THE PRESENT DAN MASA DEPAN

NEGARA-OF-THE-ART REVIEW

The Penuaan Kardiovaskular Sistem


Memahami Ini di Seluler dan Tingkat Klinis

Francesco Paneni, MD, P H D, Sebuah . b Candela Diaz Canestro, MS C, Sebuah Peter Libby, MD, P H D, c Thomas F. Luscher, MD, Sebuah . b

Giovanni G. Camici, P H D Sebuah . b

ABSTRAK

Penyakit kardiovaskular (CVD) menyajikan beban besar untuk pasien usia lanjut, pengasuh mereka, dan sistem kesehatan. perubahan struktural dan fungsional

pembuluh menumpuk sepanjang hidup, yang berpuncak pada peningkatan risiko CVD. Meningkatnya populasi lanjut usia di seluruh dunia menyoroti kebutuhan

untuk memahami bagaimana penuaan mempromosikan CVD untuk mengembangkan strategi baru untuk menghadapi tantangan ini. Ulasan ini memberikan

contoh-contoh beberapa masalah klinis yang belum terselesaikan utama yang dihadapi dalam praktek kardiovaskular sehari-hari seperti yang kita merawat pasien

usia lanjut. Berikutnya, penulis merangkum pemahaman saat ini mekanisme terlibat dalam penuaan kardiovaskular, dan potensi untuk menargetkan jalur baru

terlibat dalam disfungsi endotel, stres oksidatif mitokondria, renovasi kromatin, dan ketidakstabilan genomik. Akhirnya, penulis mempertimbangkan aspek-aspek

penting dari perbaikan pembuluh darah, termasuk transplantasi autologous sel induk berasal sumsum tulang pada pasien usia lanjut. (J Am Coll Cardiol 2017; 69:

1952 - 67) © 2017 oleh American College of Cardiology Foundation.

SEBUAH
meningkat secara dramatis, terutama di negara-negara berpenghasilan rendah dan

Penyakit (CVD) (1,2) . Memang, munculnya perawatan kontemporer menengah (misalnya, Chili, Cina, Iran, dan Rusia).

untuk akut sindrom koroner dan stroke telah membantu untuk


ge mendominasi memperpanjang
faktor risiko kardiovaskular

harapan hidup (3) . Meskipun sukses besar dari perspektif individu, resultan Meskipun penuaan hadiah array gangguan (5) .

demografi pergeseran hadiah salah satu tantangan terbesar bagi sistem sosial CVD membawa beban terbesar untuk populasi yang lebih tua, pemberi perawatan

dan perawatan kesehatan di seluruh dunia (4) . Populasi lebih dari 65 tahun akan mereka, dan sistem kesehatan (6) . Penyakit jantung koroner (PJK) asosiasi kuat

berlipat ganda dari 12% pada tahun 2010 menjadi 22% di 2040 (2) . Memang, pada dengan usia, dan merupakan penyebab utama kematian di Eropa dan Amerika

tahun 2020, jumlah orang 60 tahun dan lebih tua akan melampaui jumlah anak di Serikat (7 - 9) . Prevalensi meningkat CVD pada orang> 65 tahun, terutama pada

bawah usia 5 tahun. Laju penuaan populasi di seluruh dunia adalah mereka

> 80 tahun, dan akan meningkat w 10% selama 20 tahun ke depan (2) . Dari

2010-2030, tambahan 27 juta orang akan memiliki hipertensi, 8 juta

Dari Sebuah Pusat Molekuler Kardiologi, Universitas Zurich, Zurich, Swiss; b Universitas Heart Center, Kardiologi, Universitas Rumah Sakit Zurich, Zurich, Swiss; dan c Brigham dan Women ' s Hospital, Divisi

Kedokteran Kardiovaskular, Boston, Massachusetts. Karya ini didukung oleh Swiss National Science Foundation (Drs. Luscher dan Camici) dan Alfred dan Annemarie von Hibah Sakit untuk Translational

dan Clinical Research Kardiologi dan Onkologi (Dr. Camici), penelitian hibah tak terbatas oleh P fi zer, Inc (Dr. Luscher), Yayasan Penelitian Kardiovaskular - Zurich Heart House dan

Dengarkan naskah ini ' s audio yang

Ringkasan oleh US National Institutes of Health (RO1 HL080472), dan dari RRM Dana Amal (Dr. Libby). Drs. Paneni dan Camici adalah penerima dari Sheikh Khalifa ' Yayasan Asisten profesor di Fakultas Kedokteran
JACC Editor-in-Chief Dr Valentin Universitas Zurich. Dr Libby telah menjadi konsultan dibayar untuk Amgen, AstraZeneca, Esperion Therapeutics, Ionis Farmasi, Kowa Farmasi, Merck & Co, Novartis, P fi zer, Sano fi Regeneron, Takeda
Fuster. Farmasi, dan XBiotech; menjabat sebagai ilmiah yang fi c anggota dewan penasehat untuk Amgen, Athera Biotechnologies, Corvidia Therapeutics, DalCor Farmasi, Interleukin Genetika, Kowa Farmasi,

MedImmune, dan Novartis; dan telah menerima dana lab dari Novartis. Semua penulis lain telah melaporkan bahwa mereka tidak memiliki hubungan yang relevan dengan isi tulisan ini untuk

mengungkapkan.

Naskah diterima 12 Juli 2016; direvisi naskah diterima 23 Januari 2017, diterima 24 Januari 2017.
JACCVOL. 6 9, NO. 1 5, 2 0 1 7 Paneni et al. 1953

1 Apr 8, 2 0 1 7: 1 9 5 2 - 6 7 The Penuaan Kardiovaskular Sistem

akan memiliki PJK, 4 juta akan memiliki stroke, dan 3 juta akan memiliki gagal penentu dari kebutuhan oksigen miokard. paparan kronis meningkat SINGKATAN DAN AKRONIM

jantung akibat akumulasi cepat tua (2) . mengarah tekanan sistolik hipertrofi LV, menyebabkan kenaikan lebih

lanjut dalam permintaan oksigen miokard. Mediator, seperti TGF- b, angiotensin


BM = sumsum tulang
Peningkatan prevalensi CVD juga interplays dengan kelemahan, kondisi II, dan aldosteron mineralokortikoid,
CAC = beredar sel angiogenik
meningkatnya kerentanan terhadap stres. Sebuah meta-analisis yang termasuk

54.250 pasien usia lanjut terkait CVD dengan rasio odds (OR) dari 2,7-4,1 untuk
PJK = penyakit jantung koroner
kelemahan umum dan OR 1,5 untuk insiden kelemahan pada mereka tanpa berkontribusi secara mekanis untuk
CI = menipu fi Interval dence
kelemahan pada awal hipertrofi dan fi fibrosis di LV pressureoverloaded. Dengan demikian,
PENGEMUDI = siklooksigenase
hipertensi sistolik dan penurunan tekanan diastolik dikaitkan dengan
CV = kardiovaskular
(10) . proyeksi saat ini memprediksi peningkatan pengeluaran untuk PJK dan gagal penuaan hasil sebuah “ badai yang sempurna ” dari penurunan suplai
CVD = penyakit kardiovaskular
jantung dari w 200% selama 20 tahun ke depan, stroke diharapkan dapat oksigen dalam menghadapi kebutuhan oksigen augmented. Sebagai

memberikan kontribusi peningkatan relatif terbesar dalam biaya kesehatan tahunan aterosklerosis koroner juga kemajuan dengan usia, pembatasan eNOS = endotel oksida nitrat sintase

dari 238% (2) . Pertimbangan-pertimbangan ini menyoroti pentingnya memahami tambahan ini untuk suplai oksigen sering senyawa kebutuhan oksigen
EPC = sel progenitor endotel
mengapa usia kontribusi krusial untuk pengembangan CVD untuk memungkinkan yang meningkat biasanya ditemui dalam sistem CV penuaan ( Gambar

kita untuk mengatasi penuaan penduduk. Ulasan ini memberikan bukti klinis dan 1 ). Selain fungsi arteri besar berubah, hipertensi kronis
HFpEF = gagal jantung dengan fraksi
eksperimental untuk mendukung link didirikan antara penuaan dan CVD. mempromosikan renovasi dari mikrovaskulatur miokard. Penebalan
ejeksi diawetkan

tunika media dari arteriol miokard bisa lebih menghamhat LV perfusi


LTL = leukosit panjang telomer
dan merusak vasomotion. Konsekuensi ini dari peningkatan sistolik

dan penurunan tekanan diastolik yang biasanya menyertai penuaan LV = ventrikel kiri

bersekongkol untuk menyebabkan iskemia miokard. Regulasi renovasi


MI = infark miokard
ASPEK KLINIS CVD pada pasien usia lanjut
arteriol di miokardium mengalami peningkatan sistolik dan penurunan
MMP = matriks
tekanan diastolik kemungkinan melibatkan jalur yang sama terlibat metaloproteinase

dalam generasi kekakuan aorta. Kami kekurangan suf fi pemahaman NF k B = faktor nuklir kappa-B
Sejumlah kondisi yang berkaitan dengan usia menyajikan tantangan khusus untuk
efisien pemicu terkait usia utama untuk patofisiologi ini
ATAU = rasio odds
perawatan klinis kardiovaskular (CV) pasien kami. Akumulasi orang lanjut usia
PBMC = sel mononuklear darah
dalam populasi kami menggarisbawahi semakin pentingnya CV penuaan untuk
perifer
praktisi.
Raas = renin-angiotensinaldosterone

sistem

Hipertensi sistolik dan memperlebar PULSA TEKANAN. Dengan penuaan, menegang aorta ROS = spesies oksigen reaktif

karena peningkatan kolagen dan elastin menurun ( Gambar 1 ). RR = risiko relatif

TGF = transforming growth factor

Augmented faktor pertumbuhan transformasi (TGF) - b

Kegiatan nikmat akumulasi kolagen pada dinding aorta. Aktivitas berbagai TNF = tumor necrosis factor

elastases, termasuk matriks metalloproteinase (MMPs), seperti MMP-9 dan proses bahwa menyumbang VSMC = sel otot polos pembuluh

MMP-12, serta berlebih dari cathepsins proteinase sistein S, K, dan L, dan elastase besar terhadap komplikasi CV dalam populasi yang menua. darah

serin proteinase neutrofil, diuraikan oleh di fl Sel-sel inflamasi, semua bisa wtTTR = -Tipe liar

transthyretin
berkontribusi untuk penipisan elastin (11) . Perubahan ini di aorta ' s matriks Untung, beberapa intervensi dapat

ekstraselular kontribusi penting kerugiannya dari distensibility. Ini meningkat meningkatkan hasil pada pasien dengan hipertensi sistolik terisolasi. The SHEP

kenaikan gaji kekakuan ulang fl gelombang ected dan meningkatkan tekanan (sistolik Hipertensi pada Usia Lanjut), Syst-Eur (Systolic Hypertension di Eropa),

sistolik. Namun tekanan diastolik cenderung menurun dengan usia. Sebagai aorta Syst-China (sistolik Hipertensi di Cina), dan penelitian lain menawarkan af

pulsa gelombang kecepatan meningkat, tekanan nadi meningkat (12) . Memang, database yang kaya fi rming kemampuan pengobatan farmakologis individu dalam

tekanan nadi merupakan faktor risiko independen untuk acara CV (13) . Terisolasi 60-an atau 70-an mereka untuk mengurangi secara substansial stroke dan

hipertensi sistolik menyumbang mayoritas hipertensi yang tidak terkontrol di kematian total, dengan bene lebih rendah fi t untuk acara jantung iskemik (16) .

Amerika lebih dari 50 tahun (14,15) . Penurunan tekanan diastolik menurun drive Menghindari asupan natrium yang berlebihan dapat memberikan tambahan,

untuk perfusi koroner yang terjadi terutama selama diastole, mendukung intervensi nonfarmakologi untuk mengontrol hipertensi pada orang tua (17,18) .

pengembangan iskemia miokard. peningkatan tekanan sistolik dengan peningkatan Beberapa telah menyuarakan keprihatinan tentang keamanan agresif menurunkan

usia ventrikel kiri (LV) afterload, utama tekanan darah pada pasien usia lanjut, terutama mereka dengan penyakit arteri

koroner bersamaan (19) . Memang, kurva J-berbentuk berkaitan hasil CV tekanan

darah mungkin berkaitan dengan ini


1954 Paneni et al. JACCVOL. 6 9, NO. 1 5, 2 0 1 7

The Penuaan Kardiovaskular Sistem 1 Apr 8, 2 0 1 7: 1 9 5 2 - 6 7

GAMBAR 1 Patofisiologi aorta-LV Dynamics di CV Sistem Penuaan

Ang II ¼ angiotensin II; BP ¼ tekanan darah; CV ¼ kardiovaskular; HFpEF ¼ gagal jantung dengan fraksi ejeksi diawetkan; HFrEF ¼ gagal jantung dengan fraksi ejeksi berkurang; LV ¼ ventrikel kiri; MMP ¼ matriks

metaloproteinase; MV O2 ¼ konsumsi oksigen miokard; TGF ¼ mengubah faktor pertumbuhan.

populasi (20 - 22) . Namun HYVET (Hipertensi di Pengadilan Sangat Lansia) dan atau menipiskan risiko CVD pada orang tua.

SPRINT (Sistolik Tekanan Darah Intervensi Trial) studi con fi rmed relatif aman dan

ef fi keampuhan pengobatan antihipertensi pada populasi yang lebih tua, meskipun Beberapa (pra) studi klinis baru-baru ini telah membentuk modi vaskular kunci fi kation

data terakhir menunjukkan mempertahankan tekanan diastolik di atas 70 mm Hg terjadi dengan penuaan (31,32) , Mengidentifikasi 2 fitur utama: umum disfungsi

pada mereka dengan penyakit arteri koroner didirikan (21 - 25) . Dokter harus endotel dan kekakuan arteri pusat. Berkenaan dengan mantan, pembuluh darah

menyesuaikan agresivitas pengobatan antihipertensi pasien geriatri secara alter penuaan fungsi endotel, sel-sel yang melapisi lumen pembuluh darah.

individual, tergantung pada status arteri koroner, kelemahan, integritas fungsi disfungsi endotel termasuk mengurangi vasodilatasi dan sifat antitrombotik, dengan

otonom, dan variabel lainnya (26) . Selain mengurangi stroke, hambatan utama peningkatan stres oksidatif dan di fl sitokin inflamasi (33 - 35) mendukung

untuk hidup mandiri dan fungsi pada pasien yang lebih tua, terapi antihipertensi aterosklerosis dan trombosis, dan predisposisi untuk CVD

dapat membatasi perkembangan penyakit demensia, seperti yang ditunjukkan

dalam sidang Syst-Eur (27) . Penurunan demensia dan kognitif penurunan akru

dengan durasi yang lebih lama pengobatan antihipertensi (28) . Kerugian asimetris

untuk menindaklanjuti individu dengan gangguan kognisi mungkin bias hasilnya

demensia dalam studi SHEP ke nol (29) . (36) . Manusia dan studi eksperimental setuju bahwa bioavailabilitas berkurang dari

oksida nitrat (NO), mediator kunci dari vasorelaxation dan proses antiatherogenic,

mendasari disfungsi endotel usia tergantung (37,38) . Mengurangi bioavailabilitas

NO dapat terjadi karena sintesis menurun atau meningkat degradasi NO. Dalam

kondisi normal, endotel nitrat oksida sintase (eNOS) menghasilkan NO dari

L- arginin di hadapan tetrahydrobiopterin kofaktor (BH4) (39) . Meskipun studi

berbeda mengenai eNOS ekspresi protein dengan usia (34,40,41) , Data terakhir

VASCULAR PENUAAN: DEFINISI, PATOFISIOLOGI, DAN DAMPAK. Perubahan dalam menunjukkan suatu perubahan yang berkaitan dengan usia dalam fungsi eNOS,

struktur dan fungsi arteri menemani penuaan, dan berkontribusi terhadap disebut sebagai eNOS uncoupling ( 42) . Efek ini atau diperoleh, setidaknya

peningkatan risiko pengembangan CVD (9,30) . Dengan demikian, memahami sebagian, dari penurunan ketersediaan BH4, sehingga gangguan NO rilis dan

mekanisme yang usia mempengaruhi pembuluh darah harus memposisikan kita peningkatan produksi yang sangat pro-oksidan

untuk mencegah
JACCVOL. 6 9, NO. 1 5, 2 0 1 7 Paneni et al. 1955

1 Apr 8, 2 0 1 7: 1 9 5 2 - 6 7 The Penuaan Kardiovaskular Sistem

anion superoksida (O 2 ) ( 33) . Selain itu, ketersediaan substrat eNOS, L- arginin, juga fungsi yang mengacukan arteri homeostasis, mendukung aktivasi kronis

dapat membatasi produksi NO. Dalam hal ini, aktivitas arginase, enzim yang pro-oksidan dan proin-

bersaing dengan eNOS untuk fl jalur inflamasi, dan pergeseran ke arah pro vasokonstriktor fi le, predisposisi untuk

CVD dan efek samping (51) .

L- arginin, meningkat dengan usia, menyediakan mekanisme yang masuk akal untuk

penurunan sintesis NO (43) . Namun demikian, sebagian besar bukti mekanistik ini Peningkatan kekakuan arteri pusat, khususnya dari aorta toraks, juga

telah muncul dari studi hewan, menyoroti kebutuhan untuk penelitian lebih mencirikan penuaan pembuluh darah manusia (52) ( Gambar 1 ). Manusia dan

manusiawi mengenai batasan usia tergantung dari NO sintesis. hewan studi menunjukkan bahwa mengurangi distensibility besar hasil arteri elastis

terutama dari hilangnya elastis fi bers, serta peningkatan kolagen (53) . Mekanis,

peningkatan terkait usia degradasi elastin mungkin dihasilkan dari kegiatan

Selain itu, penuaan dapat meningkatkan NO degradasi augmented protease dengan aktivitas elastinolytic, termasuk MMPs tertentu dan

(34) karena konsentrasi tinggi dari spesies oksigen reaktif (ROS), dimediasi, cathepsins cysteinyl, enzim yang, pada gilirannya, diatur oleh di fl mediator inflamasi

sebagian, dengan di kronis fl inflamasi, yang merupakan lingkaran setan yang

menghabiskannya NO (35) . Misalnya, usia tergantung kenaikan tumor necrosis

factor pembuluh darah dan darah-ditanggung (TNF) - Sebuah, asosiasi dengan

peningkatan ekspresi nikotinamida adenin dinukleotida fosfat (NADPH) oksidase ( Gambar

2 ). Augmented NADPH aktivitas oksidase hasil kelebihan O 2 . yang, pada gilirannya, (54,55) . Kolagen katabolisme jatuh di arteri penuaan. Akumulasi modi kolagen fi ed

bereaksi dengan NO bentuk peroxynitrite (ONOO ), Oksidan kuat terlibat dalam oleh canggih akhir glikasi-produk (AGEs)

nitrosylation enzim antioksidan dan eNOS (34) . Selain di fl sitokin inflamasi, sistem (Kondensat dari

renin-angiotensin-aldosteron (RAAS) dapat berkontribusi pada peningkatan terkait glukosa yang dihasilkan oleh reaksi nonenzimatik yang membentuk cross-link

usia di NO inaktivasi. Dengan penuaan, aktivitas Raas dan konsentrasi angiotensin antara molekul kolagen) dapat menyebabkan peningkatan terkait usia ini dalam

II, suatu RAAS efektor utama, kenaikan, meningkatkan produksi ROS dengan kolagen arteri. AGE-dimediasi cross-link dapat memberikan resistensi terhadap

mengaktifkan NADPH oksidase (44) . Pada gilirannya, agerelated peningkatan degradasi enzimatik, dan dengan demikian mengganggu collagenolysis (56) . Selain

produksi ROS dapat mempromosikan vaskular itu, peningkatan aktivitas TGF b dengan penuaan merangsang sintesis kolagen

interstitial oleh sel-sel otot polos pembuluh darah (VSMC), dan dengan demikian

menambah kekakuan arteri (57) . Juga,

peningkatan aktivitas Raas mungkin

Augment kolagen sintesis dan meningkatkan elastolysis

di fl inflamasi (45) . Memang, hidrogen (58) .

peroksida (H 2 HAI 2) mengaktivasi faktor nuklir kappa-B (NF k B), yang menambah disfungsi endotel dan kekakuan arteri erat berhubungan mediator disfungsi

transkripsi proin- vaskular usia tergantung (59) . The kaku arteri, semakin besar pemaparan dari

fl gen inflamasi, yang mengarah ke peningkatan ekspresi TNF Sebuah, interleukin 6, endothelium untuk hemodinamik

kemokin, dan molekul adhesi terlibat dalam aterogenesis ( Gambar 2 ).

beban, mempromosikan endotel pengaktifan,

Peningkatan tonus vasokonstriktor mungkin senyawa gangguan agedependent di fl inflamasi, dan kerusakan (60) . pengurangan berhubungan dengan usia pada NO

di vasodilatasi endotel-dependen. Manusia penuaan rekan dengan peningkatan bioavailabilitas dijelaskan sebelumnya dapat berkontribusi untuk gangguan

sirkulasi dan tingkat dinding arteri endotelin (ET) -1, vasokonstriktor kuat terlibat distensibility arteri melalui perubahan tonus otot polos (61) . terapi baru yang

dalam gangguan dilatasi endotelium-dependent bertujuan untuk menghaluskan CVD terkait usia dapat menargetkan fitur ini

penuaan pembuluh darah, dengan penekanan khusus pada disfungsi endotel dan

perubahan dalam matriks ekstraselular arteri yang menyebabkan arteri pusat

(40,46) . Selain itu, bukti dari sel endotel tikus menunjukkan bahwa konsentrasi kekakuan.

siklooksigenase (COX) eikosanoid -derived juga berubah drastis dengan usia.

Dengan usia, eikosanoid COX-diturunkan berhubungan dengan vasokonstriksi dan

peningkatan trombosis (yaitu, prostaglandin H 2 [ PGH2], tromboksan A 2


GAGAL JANTUNG DENGAN dilestarikan dan BERKURANG fraksi ejeksi. Terkait etiologi

untuk kedua akuisisi kekakuan aorta dan renovasi dari miokardium, matriks

[TXA 2]. dan prostaglandin F 2 Sebuah [ PGF 2 Sebuah]), sedangkan prostaglandin ekstraseluler, serta mikrovaskulatur nya, gagal jantung dengan fraksi ejeksi

Saya 2 ( PGI 2, prostasiklin) jatuh (47,48) . Selain itu, gangguan respon endotel diawetkan (HFpEF) tulah pasien usia lanjut, terutama perempuan. Risiko

tergantung ke prostaglandin menyertai penuaan pada manusia dan hewan (49,50) . mengembangkan skyrockets HFpEF dengan usia (62,63) . Memang, hipertrofi

Dengan demikian, asosiasi dengan terpadu penurunan endotel penuaan


1956 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

GAMBAR 2 Representasi skematik Persiapan Molekuler Terlibat dalam arteri Aging

sel endotel berusia down-mengatur Jund dan SIRT1expression, yang mengarah ke pro-oksidan dan proin fl ekspresi gen inflamasi. Peningkatan ROS dan di fl sitokin inflamasi mengurangi ketersediaan NO. Secara paralel,

yang berkaitan dengan usia up-regulasi angiotensin II dan COX-diturunkan eikosanoid hasil mediator vasokonstriktor augmented, seperti ET-1, TXA2, dan PGF 2 Sebuah. Ketidakseimbangan antara dilator dan konstriktor

faktor petunjuknya gangguan fungsi pembuluh darah. Selanjutnya, peningkatan MMP ekspresi perubahan VSMC menginduksi dalam komponen struktural dinding arteri (misalnya, penurunan elastin / kolagen rasio),

memberikan kontribusi untuk kekakuan arteri. A A ¼ asam arakidonat; ALDH-2 ¼ aldehida dehidrogenase; AMPK ¼ 5 0 adenosine kinase protein monofosfat-diaktifkan; Arg II ¼ arginase II; AT-1 ¼ angiotensin II tipe reseptor

1; BH4 ¼ tetrahydrobiopterin; ca 2 þ ¼ ion kalsium; cGMP ¼ siklik guanosin monofosfat; PENGEMUDI ¼ cyclooxygense; DP ¼

prostaglandin reseptor D2; ecSOD ¼ ekstraseluler superoxide dismutase; EPI-4 ¼ prostaglandin E2 reseptor 1-4; ETA ¼ endotelin reseptor A; FP ¼ prostaglandin reseptor F; GC ¼ guanylate adenilat; GTP ¼ guanosin

trifosfat; ICAM ¼ molekul adhesi antar; AKU P ¼ reseptor prostasiklin; MCP ¼

monocyte chemotactic protein; MnSOD ¼ manganese superoxide dismutase; NF- k B ¼ nuclear factor k B; NO ¼ nitric oxide; PGD 2 ¼ prostaglandin D 2; PGF 2 a ¼

prostaglandin F 2 a; PGI 2 ¼ prostacyclin; PKG ¼ protein kinase G; ROS ¼ reactive oxygen species; TNF- a ¼ tumor necrosis factor alpha; TP ¼ thromboxane receptor; TxA 2 ¼ thromboxane A 2; VCAM ¼ vascular cell

adhesion protein; VSMC ¼ vascular smooth muscle cell; other abbreviations as in Figure 1 .
JACCVOL.69,NO.15,2017 Paneni et al. 1957

APRIL18,2017:1952–67 The Aging Cardiovascular System

and fi brosis of the aged LV can impair lusitropy. Practitioners generally people over 80 years of age have wtTTR amyloid deposits in their hearts at

acknowledge the increasing prevalence of HFpEF, its accentuation in elderly autopsy. Although the clinical signi fi cance of this fi nding remains uncertain, it is

patients, and its adverse outcomes in our aging patient population, with regard, not highly likely that with the aging of the population and the advent of novel diagnostic

only to morbidity, but also to reduced quality of life and increased resource modalities, wtTTR amyloidosis will become better recognized as a contributor to

utilization (64) . Unfortunately, we possess few (if any) evidence-based interventions heart failure in the older population. Amyloidosis due to wtTTR requires

to stem the development or consequences of HFpEF. Studies in progress of differentiation from amyloid light-chain amyloidosis, because the prognosis and

neurohumoral blockade may help to fi ll this important gap in our therapeutic treatment diverge substantially. Although we currently lack effective therapy for

approach to HFpEF in our elderly patients (65) . However, myocardial treating amyloidosis, promising imaging technologies should provide evolving tools

to evaluate novel therapies currently in development or under investigation (72) .

ischemia, due to either myocardial

infarction (MI) (ischemic cardiomyopathy) or subendocardial ischemia in the

hypertrophied LV, can predispose to heart failure with reduced ejection fraction.

Thus, aging can promote the development of both major forms of heart failure that
FRAILTY AND SARCOPENIA IN THE AGING POPULATION. Frailty and loss of muscle
constitute a key challenge to quality of life of aging individuals, as well as an
mass and function (sarcopenia) present a major challenge to maintenance of
enormous drain on health care resources ( Figure 1 ).
independence, mobility, quality of life, and the ability of our CV patients to undergo

surgery and other interventions (such as transcatheter valve procedures)

successfully. Moreover, the ability to tolerate CV therapies may decline with frailty

and sarcopenia, as discussed earlier for pharmacological treatment of

hypertension. We lack definite criteria for de fi ning sarcopenia and frailty


VALVULAR AND CARDIAC SKELETON CALCIFICATION. With age, calcium in the axial

skeleton particularly tends to decline, while accumulating in CV structures.

Appreciation has increased that conditions such as calci fi c aortic stenosis actually

re fl ect a systemic process (66) . Evidence points to in fl ammation as a potential

physiological contributor to CV calci fi cation. Strong human genetic data also


(73,74) , which associate with both osteoporosis and obesity (75) . Potential
implicate lipoprotein(a) in the pathogenesis of aortic valve calci fi cation (67) . Despite
pathophysiological pathways that link these apparently disparate conditions include
initial enthusiasm, clinical trials have not substantiated that statin therapy can limit
proin fl ammatory cytokines. Further clarity in diagnosis and pathophysiological
progression of aortic valvular calci fi cation. Indeed, statins tend to accelerate
mechanisms would be highly desirable, given the increase in the elderly population
coronary arterial calci fi cation (68,69) . Given the pressing clinical importance of calci fi
affected by CV disease, and the limitations these conditions present to patients and
c aortic stenosis in geriatric patients, we urgently need a greater understanding of
their management. Increased physical activity could provide a nonpharmacological
its pathophysiology and preventive measures to mitigate its genesis, as once CV
preventive approach to augmenting patient quality of life and function, and could
structures have calci fi ed, therapeutic reversal of this process may prove elusive.
conceivably improve the ability to tolerate pharmacological and interventional

treatment of CV conditions, hence improving CV outcomes.

MOLECULAR FEATURES OF AGE-RELATED

AMYLOIDOSIS AND THE AGING CV SYSTEM. Certain forms of amyloidosis also CVD

particularly affect the older population (70) . The incidence of amyloid light-chain

amyloidosis rises in older patients, related to the increase in multiple myeloma with TELOMERES AND CELLULAR SENESCENCE. Accumulation of senescent cells within

age. Cardiac amyloidosis associated with wild-type transthyretin (wtTTR) the vascular wall and heart can contribute to structural and functional decline of the

particularly affects the older population, especially men. Although many have CV system with age (58) . Considerable evidence implicates telomere shortening in

regarded cardiac amyloidosis as a rare condition, current data using novel imaging cellular senescence. Telomeres consist of repetitive nucleotide sequences

modalities show that some 13% of cases of HFpEF in individuals $60 years of age (TTAGGG) at the ends of mammalian chromosomes, that preserve chromosome

associate with wtTTR amyloidosis (71) . At least 20% of stability and integrity by preventing deterioration or fusion with neighboring

chromosomes (76) ( Central Illustration ).


1958 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

CENTRAL ILLUSTRATION Molecular Hallmarks of CV Aging (Cellular Senescence, Genomic Instability, Chromatin Remodeling, and Mitochondrial Oxidative Stress)

Paneni, F. et al. J Am Coll Cardiol. 2017;69(15):1952 – 67.

These molecular events are strongly interconnected and contribute to vascular and cardiac dysfunction in elderly patients. A thorough understanding of these complex processes may offer pharmaceutical targets to

improve human health during aging. CV ¼ cardiovascular; CVD ¼ cardiovascular disease; eNOS ¼ endothelial nitric oxide synthase; NADPH ¼ nicotinamide adenine dinucleotide phosphate; SIRT1 ¼ sirtuin 1; VSMC ¼ vascular

smooth muscle cell.

Each cell division shortens telomeric DNA until, at a critical length, the cells lose setting of CVD. Leukocyte telomere length (LTL) associates signi fi cantly with

capping function at the chromosomal ends, activating DNA damage checkpoints, vascular cell senescence, aortic valve stenosis, CV risk factors (i.e., hypertension,

cell senescence, and eventually apoptosis. Telomere shortening has particular type 2 diabetes, obesity, and smoking), and risk of atherothrombotic events.

relevance in the However, the causality of


JACCVOL.69,NO.15,2017 Paneni et al. 1959

APRIL18,2017:1952–67 The Aging Cardiovascular System

these associations remains uncertain (77) . Patients with clinical and subclinical sarcoplasmic reticulum calcium, as well as calcium release-induced I Ca calcium

features of atherosclerosis display reduced LTL compared with healthy controls, channel inactivation (84) . These changes dampen mechanical ef fi ciency and

even after adjustment for relevant confounders, such as age, sex, and race (78) . In electrophysiological properties, and increase the risk of arrhythmias (i.e., atrial fi brillation)

a recent case-control study, individuals with shorter LTL had a higher presence of in the older population.

ischemic (OR: 1.37; 95% con-

fi dence interval [CI]: 1.06 to 1.77) and hemorrhagic stroke (OR: 1.48; 95%CI: 1.08 MITOCHONDRIAL OXIDATIVE STRESS. Mitochondrial overproduction of ROS likely

to 2.02) as comparedwith the highest tertile of telomere length (79) . Moreover, contributes to cellular senescence (9) . This process ultimately leads to formation of

patients with reduced LTL had a signi fi cantly increased risk for both incident plaque the highly reactive products O 2 or H 2 O 2,

(hazard ratio:

whose accumulation and diffusion fosters senescence, DNA mutations, in fl ammation,

1.49; 95% CI: 1.09 to 2.03) and plaque progression (hazard ratio: 1.61; 95% CI: and multiple cell death pathways ( Central Illustration ) ( 82) . The mitochondrial

1.26 to 2.07) (80) . A recent meta-analysis, including prospective and retrospective

studies on the association between LTL and CHD (43,725 participants of whom adaptor p66 Shc gained

8,400 had CVD), revealed that patients with the shortest LTL had a higher relative increasing attention due to its pivotal role in ROS generation and cellular apoptosis

risk (RR) for CHD (RR: 1.54; 95%CI: 1.30 to 1.83) and cerebrovascular disease ( Central Illustration )

(RR: 1.42; 95% CI: 1.11 to 1.81) (81) . (83) . Cells lacking the p66 Shc gene have reduced intracellular free radicals, and
mice lacking p66 Shc

exposed to high oxidative stress have diminished systemic and intracellular ROS (83)

. Additionally, genetic deletion of p66 Shc in the mouse extended lifespan by 30% (84)

Critical aspects associated with cellular senescence include age-dependent . Age-dependent alteration of

defects of adrenergic

signaling and calcium handling. Plasma levels of norepinephrine signi fi cantly p66 Shc signaling profoundly affects CV homeostasis. Indeed, deletion of p66 Shc in

increase with age, as the result of reduced plasma clearance and increased mice protects from systemic and cerebral age-dependent endothelial dysfunction by

spillover from the tissues (81) . A reduction of the catecholamine reuptake virtue of decreased ROS production and conserved NO bioavailability (83,85,86) .

transporter localized in sympathetic nerve terminals also contributes to elevated Our own recent work also underscored the relevance of this gene in the

catecholamine concentrations with aging. Together, these alterations progressively pathogenesis of stroke.

impair adrenergic responsiveness, resulting in b- adrenergic desensitization. This

phenomenon ultimately reduces the number, af fi nity, and internalization of In fact,

p66 Shc- de fi cient mice display decreased ROS production in the brain, and have

reduced stroke size following ischemia-reperfusion brain injury (87) . We further

demonstrated that even post-ischemic in vivo silencing of p66 Shc prevents

ischemia-reperfusion brain injury in mice, mainly through preservation of

b- adrenergic receptors (namely the b 1-adrenergic receptor subtype), coupled with blood-brain barrier integrity (88) . The observation that p66 Shc expression increases

abnormalities in membrane adenylate cyclase activity or cellular production of cyclic signi fi cantly in stroke patients and correlates with neurological deficits supports the

adenosine monophosphate (82) . Such defects of autonomic modulation favor clinical relevance of these experimental fi ndings (88) . Expression of p66 Shc also

chronotropic incompetence and reduced inotropic reserve of the LV, thus affecting increases in peripheral blood mononuclear cells (PBMCs) of patients with acute

exercise tolerance. coronary syndrome and type 2 diabetes (89,90) . The activation of this protein by

several CV risk factors, such as hyperglycemia, oxidized low-density lipoprotein,

smoking, and hypertension, further supports its contribution to clinical CVD (9,83) .

Reduction of calcium reuptake by the myocardial sarcoplasmic reticulum Taken together, experimental and clinical data strongly indicate p66 Shc as a

calcium adenosine triphosphatase (SERCA2a) is another key feature of potential therapeutic target in the setting of age-related CVD ( Figure 2 ).

cardiomyocyte aging, yielding impaired early diastolic LV fi lling and a compensatory

increase in atrial contraction (83) . Calcium transient amplitude decreases gradually

with age, being 3.2-fold smaller in myocytes from patients $75 years of age than in

those younger than 55 years of age (84) . An age-related delay in propagation of the

calcium transient from the sarcolemma to the cell center may also occur (84) .

Moreover, aged myocytes have reduced SERCA2 expression,

The Activated Protein-1 (AP-1) transcription factor JunD has emerged as a

mediator of oxidative stress in aging ( Figure 2 ). Dimeric complexes from 3 main

limiting the amount of releasable


1960 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

families of DNA-binding proteins (Jun, Fos, and ATF/CREB) assemble to form AP-1 regulator of p66 Shc transcription, controlling acetylation of histone H3 bound to the p66

transcription factors with activities that depend critically on their speci fi c Shc promoter (102) . Impaired SIRT1 activity also favors acetylation of NF- k B p65,

components and the cellular microenvironment (91) . JunD regulates cell growth and leading to its nuclear translocation and transcription of in fl ammatory genes, as

survival, and protects cells against oxidative stress by modulating genes involved in described earlier (103) . Moreover, SIRT1 represses detrimental pathways of arterial

antioxidant defense and ROS production (92) . We recently reported that JunD falls aging, such as Forkhead box O (FOXO) 1, 3, and 4, thus preventing DNA damage,

during aging both in the mouse aorta and in PBMCs from old as compared with cell cycle arrest, and oxidative stress (104) . SIRT1 deacetylates LKB1, leading to

young, healthy humans activation of the fi nal effector enzyme 5 0 adenosine monophosphate-activated

protein kinase, a central energy regulator involved in glucose homeostasis,

maintenance of cellular adenosine triphosphate levels, and endothelial integrity via

regulation of eNOS activity and autophagy ( Figure 2 ) ( 105) .

(93) . Young mice lacking JunD display premature endothelial dysfunction and

vascular senescence similar to aged wild-type mice (93) . Aortas from

JunD / mice have augmentation of the aging markers p53 and p16 INK4a, reduced

telomerase activity, and mitochondrial DNA damage. By contrast, in vivo

overexpression of JunD rescues vascular aging features in old mice (93) .

Mechanistic experiments revealed that age-associated reductions in JunD cause an Klotho has also emerged as an important antiaging gene (106) . Klotho protein

imbalance between oxidant (NADPH oxidase) and scavenger enzymes (namely, functions as a circulating hormone that binds to a cell-surface receptor, and

manganese superoxide dismutase and aldehyde dehydrogenase represses intracellular signals of insulin and insulinlike growth factor 1, key

molecules preserving longevity (107) . Genetic deletion of Klotho in mice associates

with premature aging features, including vascular calci fi cation, altered

calcium/phosphate metabolism, and reduced lifespan (108) . By contrast,

2), leading to early redox changes, mitochondrial dysfunction, and vascular

senescence (93) . In accord with our fi ndings, lack of JunD promotes pressure

overload-induced apoptosis of myocytes, hypertrophic growth, and angiogenesis in

the heart (94) . Ongoing screening of chemical libraries aim to identify compounds Klotho overexpression prolongs lifespan in mice and protects against age-related

capable of restoring JunD activity in the vasculature and the heart. CV and renal impairment

(109) . Of clinical relevance, higher plasma Klotho levels associate independently

with a lower likelihood of CVD in humans, whereas low serum Klotho

concentrations independently predict coronary artery disease and arterial stiffness (106)

The family of nicotinamide adenine dinucleotidedependent proteins termed . Validation of Klotho as a clinically useful biomarker and a therapeutic target to

sirtuins have an established role in human aging ( Figure 2 ) ( 9) . A recent study found counteract age-related CVD requires further study.

that endogenous SIRT1 expression in VSMCs correlated inversely with donor age (95)

. Age-related loss of SIRT1 correlated with functional de fi cits, diminished stress

response, reduced capacity for migration/proliferation, and increased senescence

GENOMIC INSTABILITY. Accumulation of genetic damage through the course of life

likely contributes importantly to aging ( Central Illustration ). Genome alterations fall

(95) . Moreover, activation of SIRT1 may promote preservation of endothelial cell into 3 main categories: 1) chemical damage to genomic DNA; 2) mutations (e.g.,

function during aging. Hypercholesterolemic mice with endothelial-speci fi c SIRT1 addition, deletion, or substitution of bits of genetic code); and

overexpression or those exposed chronically to a SIRT1 activator exhibit attenuated

atherogenesis, whereas reduced SIRT1 activity results in greater foam cell

formation and atherosclerosis (96 – 98) . The observation that immunosuppressant 3) epigenetic alterations, which modify gene activity without affecting DNA

drugs (i.e., sirolimus and everolimus) cause endothelial senescence via SIRT1 sequence. To cope with genetic lesions, organisms have evolved highly pro fi-

inhibition supports these results (99) . SIRT1 blockade also impairs eNOS

functionality, whereas its activation improves endothelial NO availability (100) . cient DNA repair systems that can, in most cases, restore the correct base pair

Moreover, microRNA-217, an endogenous SIRT1 inhibitor, triggers endothelial sequence (110) . Defects in DNA repair nonetheless occur, and can contribute to

senescence by suppressing SIRT1-dependent eNOS functionality cellular senescence and organ dysfunction (111) . Genomic instability particularly

affects the CV system

(112) . Hutchinson – Gilford progeria syndrome, characterized by massive nuclear

DNA damage, associates with premature atherosclerosis and CVD, which lead to

fatal MI or stroke by an average age of 13 years

(101) . Recent work identi fi ed SIRT1 as a master


JACCVOL.69,NO.15,2017 Paneni et al. 1961

APRIL18,2017:1952–67 The Aging Cardiovascular System

(113) . Similarly, mice with genomic instability resulting from defective nucleotide epigenetic modi fi cations caused by environmental stimuli can be inherited, thus

excision repair genes contributing to early senescent traits and CVD in young adults (121) . Chromatin

ERCC1 and XPD recapitulate aging features, such as endothelial cell senescence, modi fi cations include DNA methylation and post-translational histone modi fi cations

vascular stiffness, and hypertension, at a very young age (114) . CV aging in this ( Central Illustration ). DNA methylation, which involves the addition of a methyl group

setting results mostly from eNOS and sirtuin deregulation, as well as augmented to DNA nucleotides, represses gene transcription by affecting chromatin

NADPH oxidase accessibility to the transcriptional machinery. DNA methylation progressively

decreases with age in mice and humans, whereas the rate of demethylation

(114) . associates inversely with age (122,123) . Large-scale analysis revealed

Beyond genetic diseases, a growing body of evidence indicates that sporadic unmethylated or partially methylated CpG islands in atherosclerotic plaques and

genomic mutations accumulated across the lifetime represent a major underpinning leukocytes from both patients and atherosclerosis-prone mice (124) . Changes in

of CVD (115) . Several studies have shown the presence of DNA damage in both DNA methylation localize at the promoters of several genes, including NOS,

circulating cells of patients with atherosclerosis and in the plaques themselves (116) estrogen receptors, collagen type XV alpha 1 (COL15A1), and the vascular

. PBMCs from patients with CHD have chromosomal damage and mitochondrial endothelial growth factor receptor (VEGFR). Hence, aberrant DNA methylation

DNA deletions that correlate with disease severity (117) . Similarly, data from the during the lifetime results in altered transcription of critical regulatory genes for the

AortaGen Consortium showed that genetic variation in the nucleotide excision induction of proatherogenic and senescent cellular functions (125) . Together with

repair components (namely, a single-nucleotide polymorphism [rs2029298] in the DNA-related changes,

promoter region of the DNA damage-binding protein 2 [ DDB2] gene) associated

strongly with carotid-femoral pulse wave velocity

(114) . Genomic instability in senescent VSMCs increases phosphodiesterase type post-

1 (PDE1) expression, with subsequent impairment of NO/cyclic guanosine translational histone modi fi cations, among them methylation, acetylation,

monophosphate signaling and endothelial dysfunction. Human genetic studies ubiquitination, and phosphorylation, may cluster in different patterns to regulate

consistently reveal signi fi cant associations of PDE1A single-nucleotide chromatin architecture. Histone methylation may result in different chromatin states,

polymorphisms with diastolic blood pressure and carotid intima-media thickness (118) depending on the methylated residue and the number of methyl groups added (120) .

. Overall, extensive evidence shows that genomic damage accompanies CV aging. Methylation marks on histone H3 regulate lifespan, as well as vascular homeostasis

With age, humans can accumulate somatic mutations that give rise to

hematopoietic clones associated not only with myelodysplastic syndromes and

hematologic malignancies, but surprisingly, even more strongly with CV risk (119) .

Future research in this area should unveil new interventions aimed at preserving (126,127) . For example, histone methylation by the mammalian methyltransferase

genome stability to alleviate age-related CVD. Set7 regulates endothelial NF- k B signaling, a pivotal modulator of in fl ammation and

longevity (128,129) . In addition, methylation by Set7 fundamentally regulates key

lifespan determinants heavily involved in the modulation of CV homeostasis,

namely SIRT1, FoxO3, and p53 (130) . Set7 expression increases in PBMCs of

patients with type 2 diabetes, and signi fi cantly correlates with NF- k B – mediated in fl ammation,

oxidative stress, and endothelial dysfunction, supporting the clinical relevance of

these observations (131) . Histone deacetylation by SIRT1 may also in fl uence

age-related CV disease. Transgenic overexpression of SIRT1 improves metabolic

CHROMATIN MODIFICATIONS. Although many studies have focused on the genes that ef fi ciency and endothelial function in aged mice (103) . SIRT6 can prevent

in fl uence aging, nongenomic regulation of aging has gained increasing attention. endothelial dysfunction and atherosclerosis by epigenetic modulation of multiple

Growing evidence suggests that epigenetic modi fi cations can derail transcriptional atherosclerosis-related genes, including the proatherogenic gene, tumor necrosis

programs implicated in oxidative stress, in fl ammation, angiogenesis, and cellular factor superfamily member 4 ( TNFSF4) ( 132) . These results shed light on how

chromatin modi fi cations may regulate aging-related features, and identify


metabolism, thus fostering maladaptive pathways and features of vascular aging ( Central

Illustration ) ( 120) . Epigenetic modi fi cations acquired during life appear durable and

relatively stable, thus providing a molecular framework through which the

environment interacts with the genome to alter gene expression.

Indeed,
1962 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

reversal of epigenetic modi fi cations as an attractive therapeutic target. studies have documented a reduced number of BMderived cells and CACs in aged

patients, as well as in subjects exposed to modi fi able CV risk factors

AGE-ASSOCIATED DEFECTS IN VASCULAR REPAIR (139) . Features of senescence, such as telomere shortening, genomic instability,

and subsequent cell cycle arrest, accompany this reduced cell number

IMPAIRMENT OF ANGIOGENESIS. Elderly individuals not only have increased (140) ( Figure 3 ). Kushner et al. (141) showed an approximately 60% decline in
incidence of stroke, peripheral artery disease, and MI, but also display worsened telomerase activity in CACs of older men (56 to 67 years of age). Overexpression of
outcomes than younger patients. Aged patients with acute limb ischemia have human telomerase reverse transcriptase in CACs preserved telomerase activity,
higher mortality and an increased rate of limb amputation (133) . Moreover, about delayed cell senescence, and improved age-related CAC dysfunction in mice with
35% to 40% of elderly patients experience inadequate myocardial reperfusion ischemic hind limbs (142) . Increased production of ROS and decreased expression
post-MI, eventually leading to adverse LV remodeling, heart failure, and CV death (134) of antioxidant enzymes also produces senescent cell changes. Early endothelial

. Available evidence strongly indicates an association of aging with several defects progenitor cells (EPCs) from elderly subjects show reduced activity of the
in angiogenesis. Elderly men display reduced capillary density, which is associated antioxidant enzyme glutathione peroxidase-1 in association with enhanced cell
with microvascular disease, defective eNOS functionality, and impaired insulin apoptosis (143) . Increased angiotensin II may also stimulate ROS production and
sensitivity (135) . Furthermore, senescent endothelial cells have lower proliferative cellular senescence in early EPCs
capacity, decreased telomerase activity, and reduced production of angiogenic

growth factors, such as VEGF-A. Endothelial migration is also impaired, leading to

reduced tube formation (133) . Reduced hypoxiainducible factor 1 a ( HIF1 a) activity,

mainly due to increased degradation and reduced nuclear translocation by importin a,

signi fi cantly contributes to age-related impairment of

(144) . The mobilization of progenitor cells from the BM, and their homing and
engraftment depend upon a number of angiogenic factors, SDF1/CXCR4
interaction, and VEGF (145) . We recently showed profound dysregulation of the
aging and longevity genes p66 Shc

and JunD in old (as compared with young) EPCs, promoting mitochondrial oxidative

stress and impairing SDF-1 secretion (146) . This study indicates that altered gene

expression trajectories during life strongly affect stem and progenitor cell functions.
angiogenesis after

ischemia. Interestingly, adenoviral delivery of a constitutively active form of HIF1 a improved

neovascularization and rest pain in patients with critical limb ischemia, whereas it
ROLE OF EPIGENETICS. Time-dependent accumulation of adverse epigenetic modi fi cations
failed to improve walking performance in patients with intermittent claudication (136,137)
has emerged as a potential underpinning of stem cell dysfunction in aging ( Figure 3 ).
. The activity of PGC-1 a, an emerging transcriptional coactivator that plays an
Transfer of epigenetic information (i.e., noncoding RNAs [ncRNAs]), which may
essential role in hypoxia-driven angiogenesis, also declines with age (133) .
occur either by direct cell-to-cell contact or from the bloodstream into the cell, may
Dysregulation of angiogenic pathways is associated with an age-dependent
strongly in fl uence phenotype and cell fate decisions (139) . More recent studies
reduction in the number and functionality of stem and progenitor cells (i.e.,
showed that extracellular vesicles, including microparticles, exosomes, and
circulating angiogenic cells [CACs] and bone marrow [BM]-derived cells) (133) .
apoptotic bodies, can mediate intercellular communication of BM-derived cells with
Understanding how aging deteriorates stem cell functionality is of paramount
other cell types, with modi fi cation of the transcriptional pro fi le mediated by ncRNAs (147)
importance to develop new therapeutic approaches in this area.
. MicroRNA pro fi ling in BM cells from young and aged mice revealed that miR-10A*

and miR-21, and their common target gene Hmga2, can drive EPC senescence (148)

. Suppression of those microRNAs in aged EPCs increased Hmga2 expression and

blunted expression of the senescence genes p16 INK4a/ p19 ARF,

MECHANISMS OF STEM CELL AGING. Recent data obtained in hematopoietic stem

cells (HSCs) indicate that, by the age of 70 years, clonal diversity collapses,

resulting in the dominance of 1 HSC clone (138) . Such collapse of highly polyclonal

into quasi-monotypic hematopoiesis may in fl uence stem cell competency during

ischemia or infarction. In addition, several thus ameliorating angiogenic properties. In another study, miR-34a induced EPC

senescence by suppressing SIRT1 (149) . These novel data illustrate how


JACCVOL.69,NO.15,2017 Paneni et al. 1963

APRIL18,2017:1952–67 The Aging Cardiovascular System

FIGURE 3 Features of Age-Related Stem Cell Dysfunction and Approaches to Boost Their Therapeutic Potential in Elderly Patients With CVD

elderly patient with


CVD (MI, HF)
epigenetic changes
“ Dysfunctional
BM-derived cells ” genomic instability

clonal diversity

telomere shortening

oxidative stress
“aged” bone marrow
inflammation

ex-vivo reprogramming in situ reprogramming


(epigenetic signals, gene therapy) (cardiomyocyte dedifferentiation, stimulation
of endogenous CSC)

intracoronary stem cell reinfusion

Future strategies to ameliorate the ef fi ciency of autologous transplantation may include ex vivo reprogramming of maladaptive pathways, in situ reprogramming by stimulating

cardiomyocyte dedifferentiation, or in situ stimulation of endogenous cardiac stem cells. BM ¼ bone marrow; CSC ¼ cardiac stem cell; CVD ¼ cardiovascular disease; HF ¼ heart failure;

MI ¼ myocardial infarction.

editing the epigenetic landscape of aged EPCs can furnish a novel the SWISS AMI trial (Swiss Multicenter Intracoronary Stem Cells Study in Acute

mechanism-based approach to promoting vascular repair in elderly patients with Myocardial Infarction), which recruited a similar number of patients, treatment with

CVD. BM-derived mononuclear cells for either 5 to 7 days or 3 to 4 weeks after acute MI

failed to improve LV function (assessed by cardiac magnetic resonance imaging) at

STEM CELL – BASED THERAPIES. Available evidence on the CV effects of stem 12 months (152) . Furthermore, a very recent meta-analysis in patients with

cell-based therapies is overall very dif fi cult to interpret due to heterogeneous refractory angina showed that cell-based therapy improves anginal episodes,

populations, and different technical

approaches. The ACCRUE study (meta-Analysis of Cell-based CaRdiac stUdiEs),

which included randomized trials in patients with a recent acute MI, revealed that reduces the use of

intracoronary cell therapy provided no bene fi t, in terms of clinical events or changes antianginal medications, ameliorates exercise tolerance and myocardial perfusion,

in LV function (150) . However, the PreSERVE-AMI trial (A Prospective Randomized and reduces the risk of major adverse cardiac events and arrhythmias compared

Double Blind Placebo Controlled Phase II Trial of Intra-coronary Infusion of with maximal medical therapy (153) . The explanation behind these con fl icting and

AMR-001, a Bone Marrow Derived Autologous CD34 þ highly debated results lies in the fact that, in most cases,

“ aged ” BM cells, which have already traveled down the aforementioned senescence

Selected Cell Product, in Patients With Acute pathways, are reinfused. Moreover, the older recipient organ presents a diseased

Myocardial Infarction), the largest study of cell-based therapy for ST-segment milieu whereby activation of several pathophysiological pathways may hamper the

elevation MI completed in the United States, showed that intracoronary infusion of healing potential of stem cells. In this regard, new-generation approaches should

autologous CD34 þ cells in patients with ischemic LV dysfunction may exert include identi fi cation of competent cells from the BM aspirate, as well as ex vivo

favorable, albeit small, cell dose-dependent effect on LV ejection fraction, infarct reprogramming of maladaptive pathways and

size, and survival after discharge (151) . By contrast, in


1964 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

epigenetic signals. Of clinical relevance, the growing understanding of chromatin JunD, and SIRT1, have gained increasing attention due to their involvement in

architecture and metabolism has led to the design of speci fi c compounds (some essential pathways regulating ROS production and/or scavenging and proin fl ammatory

already approved by the Food and Drug Administration and tested in clinical trials) cytokines. Indeed, the SIRT1 activator SRT2104 has already undergone clinical

that can modulate epigenetic modi fi cations, and thereby restore gene expression. study

(157) . These more targeted and selective interventions merit attention, given the

disappointments encountered with the application of blunt approaches to limit

oxidative stress in humans. Recent clinical trials have shown that therapeutic
CONCLUSIONS AND FUTURE PERSPECTIVES
targeting of oxidative stress using ROS scavengers not only lacks ef fi cacy, but can

even prove harmful (82) . Ultimately, from a lifestyle perspective, regimens such as
More than 2 decades of dedicated research have
caloric restriction or regular endurance exercise may attenuate signs of vascular
fi rmly established the concept that increased oxidative stress and in fl ammation
aging by increasing SIRT1 levels, PGC-1 a – dependent mitochondrial biogenesis,
promote CV aging. However, generalized antioxidant supplementation, including
eNOS functionality, and antioxidant response via enhanced activity of the
vitamin E and b- carotene, failed to reduce CV events in asymptomatic individuals,
transcription factor Nfr-2 (41,158) . Translation of the basic and clinical science
as well as in patients at high CV risk (154,155) . Anti-in fl ammatory interventions,
reviewed here should prepare us better to confront the burden of CVD in our
such as anti-TNF a treatments, did not demonstrate bene fi cial effects on morbidity
growing older population.
and mortality in patients with chronic heart failure

(156) . Despite these disappointing results, emerging innovative strategies that

target ROS, either by inhibiting speci fi c pro-oxidant enzymes or by augmenting

endogenous antioxidants, have yielded some promising preclinical results. Other

efforts seek to develop selective anti-in fl ammatory agents that might reduce CVD

without disturbing metabolic homeostasis. New molecular targets, such as p66 Shc, ADDRESS FOR CORRESPONDENCE: Prof. Giovanni G. Camici, Center for Molecular

Cardiology, University of Zurich, Wagistrasse 12, Schlieren CH-8952, Zurich,

Switzerland. E-mail: giovanni.camici@uzh.ch .

RE F E RENCE S

1. North BJ, Sinclair DA. The intersection between aging and cardiovascular 8. Newman AB, Naydeck BL, Ives DG, et al. Coronary artery calcium, 15. Chobanian AV. Clinical practice. Isolated systolic hypertension in the

disease. Circ Res 2012; 110:1097 – 108 . carotid artery wall thickness, and cardiovascular disease outcomes in adults elderly. N Engl J Med 2007;357:789 – 96 .

70 to 99 years old. Am J Cardiol 2008;101:186 – 92 .

2. Heidenreich PA, Trogdon JG, Khavjou OA, et al. Forecasting the future of 16. Staessen JA, Gasowski J, Wang JG, et al. Risks of untreated and

cardiovascular disease in the United States: a policy statement from the 9. Camici GG, Savarese G, Akhmedov A, et al. Molecular mechanism of treated isolated systolic hypertension in the elderly: meta-analysis of

American Heart Association. Circulation 2011;123: 933 – 44 . endothelial and vascular aging: implications for cardiovascular disease. Eur outcome trials [Published correction appears in Lancet 2001;357:724].

Heart J 2015;36:3392 – 403 . Lancet 2000;355:865 – 72 .

10. A fi lalo J, Karunananthan S, Eisenberg MJ, et al. Role of frailty in 17. Appel LJ, Espeland MA, Easter L, et al. Effects of reduced sodium
3. Fleg JL, Aronow WS, Frishman WH. Cardiovascular drug therapy in the
patients with cardiovascular disease. Am J Cardiol 2009;103:1616 – 21 . intake on hypertension control in older individuals: results from the Trial of
elderly: bene fi ts and challenges. Nat Rev Cardiol 2011;8:13 – 28 .
Nonpharmacologic Interventions in the Elderly (TONE). Arch Intern Med

2001;161:685 – 93 .
11. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology,
4. Christensen K, Doblhammer G, Rau R, et al. Ageing populations: the
and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol 2005;25: 932 –
challenges ahead. Lancet 2009;374:1196 – 208 .
43 .
18. Cook NR, Appel LJ, Whelton PK. Sodium intake and all-cause mortality

over 20 years in the Trials of Hypertension Prevention. J Am Coll Cardiol


5. Freedman VA, Martin LG, Schoeni RF. Recent trends in disability and 2016;68:1609 – 17 .
12. McEniery CM, Wilkinson IB, Avolio AP. Age, hypertension and arterial
functioning among older adults in the United States: a systematic review.
function. Clin Exp Pharmacol Physiol 2007;34:665 – 71 .
JAMA 2002;288:3137 – 46 .
19. Messerli FH, Mancia G, Conti CR, et al. Dogma disputed: can

13. Franklin SS, Khan SA, Wong ND, et al. Is pulse pressure useful in aggressively lowering blood pressure in hypertensive patients with coronary

6. Kovacic JC, Moreno P, Nabel EG, et al. Cellular senescence, vascular predicting risk for coronary heart disease? The Framingham Heart Study. artery disease be dangerous? Ann Intern Med 2006;144: 884 – 93 .

disease, and aging: part 2 of a 2-part review: clinical vascular disease in the Circulation 1999;100:354 – 60 .
elderly. Circulation 2011;123:1900 – 10 .

14. Franklin SS, Jacobs MJ, Wong ND, et al. Predominance of 20. Vidal-Petiot E, Ford I, Greenlaw N, et al. Cardiovascular event rates and

7. Kuller LH, Lopez OL, Mackey RH, et al. Subclinical cardiovascular isolated systolic hypertension mortality according to achieved systolic and diastolic blood pressure in

disease and death, dementia, and coronary heart disease in patients 80 þ years. among middle-aged and elderly US hypertensives: analysis based on patients with stable coronary artery disease: an international cohort study.

J Am Coll Cardiol 2016;67: 1013 – 22 . National Health and Nutrition Examination Survey (NHANES) III. Lancet 2016;388: 2142 – 52 .

Hypertension 2001;37:869 – 74 .
JACCVOL.69,NO.15,2017 Paneni et al. 1965

APRIL18,2017:1952–67 The Aging Cardiovascular System

21. Selvaraj S, Steg PG, Elbez Y, et al., REACH Registry Investigators. 37. Tschudi MR, Barton M, Bersinger NA, et al. Effect of age on kinetics of the Atherosclerosis Risk in Communities (ARIC) Study. Am J Hypertens

Pulse pressure and risk for cardiovascular events in patients with nitric oxide release in rat aorta and pulmonary artery. J Clin Invest 1996; 2016;29:114 – 22 .

atherothrombosis: from the REACH Registry. J Am Coll Cardiol 98:899 – 905 .


53. Fritze O, Romero B, Schleicher M, et al. Agerelated changes in the
2016;67:392 – 403 .
elastic tissue of the human aorta. J Vasc Res 2012;49:77 – 86 .

38. Taddei S, Virdis A, Ghiadoni L, et al. Agerelated reduction of NO

22. McEvoy JW, Chen Y, Rawlings A, et al. Diastolic blood pressure, availability and oxidative stress in humans. Hypertension 2001;38:274 – 9 .
54. Liu J, Sukhova GK, Sun JS, et al. Lysosomal cysteine proteases in
subclinical myocardial damage, and cardiac events: implications for blood
atherosclerosis. Arterioscler Thromb Vasc Biol 2004;24:1359 – 66 .
pressure control. J Am Coll Cardiol 2016;68: 1713 – 22 .
39. Lüscher TF, Yang ZH, Diederich D, et al. Endothelium-derived

vasoactive substances: potential role in hypertension, atherosclerosis, and


55. Li Z, Froehlich J, Galis ZS, et al. Increased expression of matrix
vascular occlusion. J Cardiovasc Pharmacol 1989; 14 Suppl 6:S63 – 9 .
metalloproteinase-2 in the thickened intima of aged rats. Hypertension
23. Beckett NS, Peters R, Fletcher AE, et al., HYVET Study Group.
1999; 33:116 – 23 .
Treatment of hypertension in patients 80 years of age or older. N Engl J

Med 2008;358:1887 – 98 .
40. Donato AJ, Gano LB, Eskurza I, et al. Vascular endothelial dysfunction 56. Aronson D. Cross-linking of glycated collagen in the pathogenesis of
with aging: endothelin-1 and endothelial nitric oxide synthase. Am J Physiol arterial and myocardial stiffening of aging and diabetes. J Hypertens
24. Williamson JD, Supiano MA, Applegate WB, et al., SPRINT Research Heart Circ Physiol 2009;297:H425 – 32 . 2003;21:3 – 12 .
Group. Intensive vs standard blood pressure control and cardiovascular

disease outcomes in adults aged $75 years: a randomized clinical trial.


41. Durrant JR, Seals DR, Connell ML, et al. Voluntary wheel running 57. Amento EP, Ehsani N, Palmer H, et al. Cytokines and growth factors
JAMA 2016;315:2673 – 82 .
restores endothelial function in conduit arteries of old mice: direct evidence positively and negatively regulate interstitial collagen gene expression in
for reduced oxidative stress, increased superoxide dismutase activity and human vascular smooth muscle cells. Arterioscler Thromb 1991;11:1223 – 30
25. Bhatt DL. Troponin and the J-curve of diastolic blood pressure: when
downregulation of NADPH oxidase. J Physiol 2009; 587:3271 – 85 . .
lower is not better. J Am Coll Cardiol 2016;68:1723 – 6 .

58. Huang W, Alhenc Gelas F, OsbornePellegrin MJ. Protection of the


26. Fuster V. No such thing as ideal blood pressure: a case for personalized
arterial internal elastic lamina by inhibition of the reninangiotensin system in
medicine. J Am Coll Cardiol 2016;67:3014 – 5 .
42. Yang YM, Huang A, Kaley G, et al. eNOS uncoupling and endothelial the rat. Circ Res 1998;82: 879 – 90 .

dysfunction in aged vessels. Am J Physiol Heart Circ Physiol 2009;297:

27. Forette F, Seux ML, Staessen JA, et al. Prevention of dementia in H1829 – 36 .

randomised double-blind placebo-controlled Systolic Hypertension


59. Jani c M, Lunder M, Sabovi c M. Arterial stiffness and cardiovascular
in
43. Santhanam L, Christianson DW, Nyhan D, et al. Arginase and vascular therapy. Biomed Res Int 2014;2014:621437 .
Europe (Syst-Eur) trial. Lancet 1998;352:1347 – 51 .
aging. J Appl Physiol (1985) 2008;105:1632 – 42 .

28. Peila R, White LR, Masaki K, et al. Reducing the risk of dementia: ef fi cacy
60. Walker AE, Henson GD, Reihl KD, et al. Greater impairments in
of long-term treatment of hypertension. Stroke 2006;37:1165 – 70 .
44. Dikalov SI, Nazarewicz RR. Angiotensin IIinduced production of cerebral artery compared with skeletal muscle feed artery endothelial

mitochondrial reactive oxygen species: potential mechanisms and function in a mouse model of increased large artery stiffness. J Physiol

29. Di Bari M, Pahor M, Franse LV, et al. Dementia and disability outcomes relevance for cardiovascular disease. Antioxidants Redox Signaling 2015;593:1931 – 43 .

in large hypertension trials: lessons learned from the systolic hypertension 2013;19:1085 – 94 .

in the elderly program (SHEP) trial. Am J Epidemiol 2001;153:72 – 8 .


61. Isabelle M, Simonet S, Ragonnet C, et al. Chronic reduction of nitric

45. Ungvari Z, Orosz Z, Labinskyy N, et al. Increased mitochondrial H2O2 oxide level in adult spontaneously hypertensive rats induces aortic stiffness

production promotes endothelial NF- k B activation in aged rat arteries. Am J similar to old spontaneously hypertensive rats. J Vasc Res 2012;49:309 – 18 .

30. Wilson PW, D ’ Agostino RB, Levy D, et al. Prediction of coronary heart Physiol Heart Circ Physiol 2007;293: H37 – 47 .
disease using risk factor categories. Circulation 1998;97:1837 – 47 .

62. Lloyd-Jones DM, Larson MG, Leip EP, et al. Lifetime risk for developing

31. Vita JA, Treasure CB, Nabel EG, et al. Coronary vasomotor response to congestive heart failure: the Framingham Heart Study. Circulation
46. Van Guilder GP, Westby CM, Greiner JJ, et al. Endothelin-1
acetylcholine relates to risk-factors for coronary-artery disease. Circulation 2002;106:3068 – 72 .
vasoconstrictor tone increases with age in healthy men but can be reduced
1990;81:491 – 7 . by regular aerobic exercise. Hypertension 2007;50:403 – 9 .

63. Red fi eld MM. Heart failure with preserved ejection fraction. N Engl J

32. Celermajer DS, Sorensen KE, Spiegelhalter DJ, et al. Aging is Med 2016;375:1868 – 77 .
47. Tang EHC, Vanhoutte PM. Gene expression changes of prostanoid
associated with endothelial dysfunction in healthy men years before the
synthases in endothelial cells and prostanoid receptors in vascular smooth 64. Owan TE, Hodge DO, Herges RM, et al. Trends in prevalence and
agerelated decline in women. J Am Coll Cardiol 1994;24:471 – 6 .
muscle cells caused by aging and hypertension. Physiol Genomics outcome of heart failure with preserved ejection fraction. N Engl J Med

2008;32:409 – 18 . 2006; 355:251 – 9 .

33. Delp MD, Behnke BJ, Spier SA, et al. Ageing diminishes
48. Heymes C, Habib A, Yang D, et al. Cyclooxygenase-1 and -2 65. McMurray JJ, Packer M, Desai AS, et al., PARADIGM-HF Committees
endothelium-dependent vasodilatation and tetrahydrobiopterin content in rat
contribution to endothelial dysfunction in ageing. Br J Pharmacol 2000;131: and Investigators. Dual angiotensin receptor and neprilysin inhibition as an
skeletal muscle arterioles. J Physiol 2008;586:1161 – 8 .
804 – 10 . alternative to angiotensin-converting enzyme inhibition in patients with

chronic systolic heart failure: rationale for and design of the Prospective

34. van der Loo B, Labugger R, Skepper JN, et al. Enhanced peroxynitrite comparison of ARNI with ACEI to Determine Impact on Global Mortality and
49. Singh N, Prasad S, Singer D, et al. Ageing is associated with
formation is associated with vascular aging. J Exp Med 2000;192: 1731 – 44 . morbidity in Heart Failure trial (PARADIGM-HF). Eur J Heart Fail
impairment of nitric oxide and prostanoid dilator pathways in the human
2013;15:1062 – 73 .
forearm. Clin Sci (Lond) 2002;102:595 – 600 .

35. Donato AJ, Eskurza I, Silver AE, et al. Direct evidence of endothelial
50. Küng CF, Lüscher TF. Different mechanisms of endothelial dysfunction
oxidative stress with aging in humans: relation to impaired
with aging and hypertension in rat aorta. Hypertension 1995;25:194 – 200 .
endotheliumdependent dilation and upregulation of nuclear factor- k B. Circ 66. Otto CM, Prendergast B. Aortic-valve stenosis —

Res 2007;100:1659 – 66 . from patients at risk to severe valve obstruction. N Engl J Med

51. Camici GG, Sudano I, Noll G, et al. Molecular pathways of aging and 2014;371:744 – 56 .

hypertension. Curr Opin Nephrol Hypertens 2009;18:134 – 7 .


36. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in 67. Thanassoulis G, Campbell CY, Owens DS, et al., CHARGE

cardiovascular disease enterprises: part I: aging arteries: a “ set up ” for Extracoronary Calcium Working Group. Genetic associations with valvular

vascular disease. Circulation 2003;107:139 – 46 . 52. Meyer ML, Tanaka H, Palta P, et al. Correlates of segmental pulse calci fi cation and aortic stenosis. N Engl J Med 2013;368:503 – 12 .

wave velocity in older adults:


1966 Paneni et al. JACCVOL.69,NO.15,2017

The Aging Cardiovascular System APRIL18,2017:1952–67

68. Libby P. How does lipid lowering prevent coronary events? New insights response and life span in mammals. Nature 1999; 402:309 – 13 . implication of cilostazol after drug-eluting stent implantation. J Am Coll

from human imaging trials. Eur Heart J 2015;36:472 – 4 . Cardiol 2009;53: 2298 – 305 .

85. Francia P, delli Gatti C, Bachschmid M, et al. Deletion of p66shc gene

69. Henein M, Granåsen G, Wiklund U, et al. High dose and long-term protects against agerelated endothelial 100. Ota H, Akishita M, Eto M, et al. Sirt1 modulates premature

statin therapy accelerate coronary artery calci fi cation. Int J Cardiol 2015; dysfunction. Circulation senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol

184:581 – 6 . 2004;110:2889 – 95 . 2007; 43:571 – 9 .

86. Shi Y, Savarese G, Perrone-Filardi P, et al. Enhanced

70. Falk RH. Diagnosis and management of the cardiac amyloidoses. age-dependent cerebrovascular 101. Menghini R, Casagrande V, Cardellini M, et al. MicroRNA 217

Circulation 2005;112: dysfunction is mediated by adaptor protein p66Shc. Int J Cardiol modulates endothelial cell senescence via silent information regulator 1.

2047 – 60 . 2014;175:446 – 50 . Circulation 2009;120:1524 – 32 .

87. Spescha RD, Shi Y, Wegener S, et al. Deletion of the ageing gene
71. González-López E, Gallego-Delgado M, GuzzoMerello G, et al.
p66Shc reduces early stroke size following ischaemia/reperfusion brain 102. Zhou S, Chen HZ, Wan YZ, et al. Repression of P66Shc expression by
Wild-type transthyretin amyloidosis as a cause of heart failure with
injury. Eur Heart J 2013;34:96 – 103 . SIRT1 contributes to the prevention of hyperglycemia-induced endothelial
preserved ejection fraction. Eur Heart J 2015;36: 2585 – 94 .

dysfunction. Circ Res 2011;109:639 – 48 .

88. Spescha RD, Klohs J, Semerano A, et al. Postischaemic silencing of

p66Shc reduces ischaemia/ reperfusion brain injury and its expression


72. Falk RH. Pondering the prognosis and pathology of cardiac amyloidosis: 103. Winnik S, Auwerx J, Sinclair DA, et al. Protective effects of sirtuins in
correlates to clinical outcome in stroke. Eur Heart J 2015;36:1590 – 600 .
answers breed questions. J Am Coll Cardiol Img 2016;9:139 – 41 . cardiovascular diseases: from bench to bedside. Eur Heart J 2015; 36:3404 –

12 .

73. Rosenberg IH. Sarcopenia: origins and clinical relevance. J Nutr


89. Franzeck FC, Hof D, Spescha RD, et al. Expression of the aging gene
1997;127:990S – 1S . 104. Brunet A, Sweeney LB, Sturgill JF, et al. Stress-dependent regulation
p66Shc is increased in peripheral blood monocytes of patients with acute
of FOXO transcription factors by the SIRT1 deacetylase. Science
74. Curtis E, Litwic A, Cooper C, et al. Determinants of muscle and bone coronary syndrome but not with stable coronary artery disease.
2004;303:2011 – 5 .
aging. J Cell Physiol 2015;230:2618 – 25 . Atherosclerosis 2012;220: 282 – 6 .

105. Mattagajasingh I, Kim CS, Naqvi A, et al. SIRT1 promotes


75. Reginster JY, Beaudart C, Buckinx F, et al. Osteoporosis and
endothelium-dependent vascular relaxation by activating endothelial nitric
sarcopenia: two diseases or one? Curr Opin Clin Nutr Metab Care 90. Pagnin E, Fadini G, de Toni R, et al. Diabetes induces p66shc gene
oxide synthase. Proc Natl Acad Sci U S A 2007;104: 14855 – 60 .
2016;19:31 – 6 . expression in human peripheral blood mononuclear cells: relationship to

oxidative stress. J Clin Endocrinol Metab 2005;90: 1130 – 6 .


76. Calado RT, Young NS. Telomere diseases. N Engl J Med

2009;361:2353 – 65 .
106. Martín-Núñez E, Donate-Correa J, Muros-deFuentes M, et al.

77. Kurz DJ, Kloeckener-Gruissem B, Akhmedov A, et al. Degenerative Implications of Klotho in vascular health and disease. World J Cardiol
91. Hernandez JM, Floyd DH, Weilbaecher KN, et al. Multiple facets of junD
aortic valve stenosis, but not coronary disease, is associated with shorter 2014;6: 1262 – 9 .
gene expression are atypical among AP-1 family members. Oncogene
telomere length in the elderly. Arterioscler Thromb Vasc Biol 2006;26:e114 – 7
2008;27:4757 – 67 .
.
107. Ding HY, Ma HX. Signi fi cant roles of antiaging protein klotho and fi broblast

growth factor23 in cardiovascular disease. J Geriatr Cardiol 2015;12:439 – 47


92. Gerald D, Berra E, Frapart YM, et al. JunD reduces tumor angiogenesis
78. Brouilette SW, Moore JS, McMahon AD, et al., West of Scotland .
by protecting cells from oxidative stress. Cell 2004;118:781 – 94 .
Coronary Prevention Study Group. Telomere length, risk of coronary heart

disease, and statin treatment in the West of Scotland Primary Prevention


108. Lindberg K, Olauson H, Amin R, et al. Arterial klotho expression and
93. Paneni F, Osto E, Costantino S, et al. Deletion of the activated protein-1
Study: a nested casecontrol study. Lancet 2007;369:107 – 14 .
FGF23 effects on vascular calci fi cation and function. PLoS One 2013;8:
transcription factor JunD induces oxidative stress and accelerates
e60658 .
agerelated endothelial dysfunction. Circulation 2013; 127:1229 – 40, e1 – 21 .

79. Zhang W, Chen Y, Wang Y, et al. Short telomere length in blood


109. Kurosu H, Yamamoto M, Clark JD, et al. Suppression of aging in mice
leucocytes contributes to the presence of atherothrombotic stroke and
by the hormone Klotho. Science 2005;309:1829 – 33 .
94. Ricci R, Eriksson U, Oudit GY, et al. Distinct functions of junD in cardiac
haemorrhagic stroke and risk of post-stroke death. Clin Sci (Lond)
hypertrophy and heart failure. Genes Dev 2005;19:208 – 13 .
2013;125:27 – 36 .
110. Lord CJ, Ashworth A. The DNA damage response and cancer therapy.

95. Thompson AM, Wagner R, Rzucidlo EM. Agerelated loss of SirT1 Nature 2012;481: 287 – 94 .
80. Chen S, Lin J, Matsuguchi T, et al. Short leukocyte telomere length
expression results in dysregulated human vascular smooth muscle cell
predicts incidence and progression of carotid atherosclerosis in American
function. Am J Physiol Heart Circ Physiol 2014; 307:H533 – 41 .
111. Vijg J, Suh Y. Genome instability and aging. Annu Rev Physiol
Indians: the Strong Heart Family Study. Aging (Albany NY) 2014;6:414 – 27 .
2013;75:645 – 68 .

112. Busuttil RA, Dollé M, Campisi J, et al. Genomic instability, aging, and
96. ZhangQJ,WangZ, ChenHZ, et al. Endotheliumspeci fi c overexpression of
cellular senescence. Ann N Y Acad Sci 2004;1019:245 – 55 .
81. Haycock PC, Heydon EE, Kaptoge S, et al. Leucocyte telomere length
class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein
and risk of cardiovascular disease: systematic review and meta-analysis.
E-de fi cient mice. Cardiovasc Res 2008;80: 191 – 9 .
BMJ 2014;349:g4227 . 113. Capell BC, Collins FS, Nabel EG. Mechanisms of cardiovascular

disease in accelerated aging syndromes. Circ Res 2007;101:13 – 26 .

82. Kornfeld OS, Hwang S, Disatnik MH, et al. Mitochondrial reactive 97. Stein S, Lohmann C, Schäfer N, et al. SIRT1 decreases Lox-1-mediated
oxygen species at the heart of the matter: new therapeutic approaches for foam cell formation in atherogenesis. Eur Heart J 2010;31:2301 – 9 . 114. Durik M, Kavousi M, van der Pluijm I, et al. Nucleotide excision DNA

cardiovascular diseases. Circ Res 2015;116: 1783 – 99 . repair is associated with age-related vascular dysfunction. Circulation

2012;126:468 – 78 .
98. Miranda MX, van Tits LJ, Lohmann C, et al. The Sirt1 activator

SRT3025 provides atheroprotection in Apoe-/- mice by reducing hepatic

83. Cosentino F, Francia P, Camici GG, et al. Final common molecular Pcsk9 secretion and enhancing Ldlr expression. Eur Heart J 2015;36:51 – 9 . 115. Shah NR, Mahmoudi M. The role of DNA damage and repair in

pathways of aging and cardiovascular disease: role of the p66Shc protein. atherosclerosis: a review. J Mol Cell Cardiol 2015;86:147 – 57 .

Arterioscler Thromb Vasc Biol 2008;28:622 – 8 .

99. Ota H, Eto M, Ako J, et al. Sirolimus and everolimus induce endothelial 116. Mahmoudi M, Mercer J, Bennett M. DNA damage and repair in

84. Migliaccio E, Giorgio M, Mele S, et al. The p66shc adaptor protein cellular senescence via sirtuin 1 down-regulation: atherosclerosis. Cardiovasc Res 2006;71:259 – 68 .

controls oxidative stress therapeutic


JACCVOL.69,NO.15,2017 Paneni et al. 1967

APRIL18,2017:1952–67 The Aging Cardiovascular System

117. Botto N, Rizza A, Colombo MG, et al. Evidence for DNA damage in 133. Lähteenvuo J, Rosenzweig A. Effects of aging on angiogenesis. Circ proangiogenic effects and alterations in type 2 diabetics. Blood

patients with coronary artery disease. Mutat Res 2001;493:23 – 30 . Res 2012;110:1252 – 64 . 2013;121:226 – 36 .

134. Zhang H, van Olden C, Sweeney D, et al. Blood vessel repair and 148. Zhu S, Deng S, Ma Q, et al. MicroRNA-10A* and microRNA-21

118. Bautista Niño PK, Durik M, Danser AH, et al. Phosphodiesterase 1 regeneration in the ischaemic heart. Open Heart 2014;1:e000016 . modulate endothelial progenitor cell senescence via suppressing

regulation is a key mechanism in vascular aging. Clin Sci (Lond) 2015;129: high-mobility group A2. Circ Res 2013;112:152 – 64 .

1061 – 75 .
135. Groen BB, Hamer HM, Snijders T, et al. Skeletal muscle capillary

density and microvascular function are compromised with aging and type 2 149. Zhao T, Li J, Chen AF. MicroRNA-34a induces endothelial progenitor

119. Jaiswal S, Fontanillas P, Flannick J, et al. Agerelated clonal diabetes. J Appl Physiol cell senescence and impedes its angiogenesis via suppressing silent

hematopoiesis associated with adverse outcomes. NEngl JMed (1985) 2014;116: information regulator 1. Am J Physiol Endocrinol Metab 2010;299:E110 – 6 .

2014;371:2488 – 98 . 998 – 1005 .

120. Costantino S, Paneni F, Cosentino F. Targeting chromatin remodeling 136. Creager MA, Olin JW, Belch JJ, et al. Effect of hypoxia-inducible

to prevent cardiovascular disease in diabetes. Curr Pharm Biotechnol factor-1 a gene therapy on walking performance in patients with intermittent 150. Gyöngyösi M, Wojakowski W,

2015;16:531 – 43 . claudication. Circulation 2011;124:1765 – 73 . Lemarchand P, et al., ACCRUE Investigators. Meta-Analysis of Cell-based

CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction

based on individual patient data. Circ Res 2015;116:1346 – 60 .


121. Paneni F, Costantino S, Volpe M, et al. Epigenetic signatures and 137. Rajagopalan S, Olin J, Deitcher S, et al. Use of a constitutively active

vascular risk in type 2 diabetes: a clinical perspective. Atherosclerosis hypoxia-inducible factor-1 a

2013;230:191 – 7 . transgene as a therapeutic strategy in no-option critical limb ischemia

patients: phase I doseescalation experience. Circulation 2007;115: 1234 – 43 . 151. Quyyumi AA, Vasquez A, Kereiakes D, et al. PreSERVE-AMI: a

randomized, double-blind, placebo-controlled clinical trial of intracoronary


122. Singhal RP, Mays-Hoopes LL, Eichhorn GL. DNA methylation in aging
administration of autologous CD34 þ cells in patients with left ventricular
of mice. Mech Ageing Dev 1987;41:199 – 210 .
dysfunction post STEMI. Circ Res 2017;120:324 – 31 .
138. Goodell MA, Rando TA. Stem cells and healthy aging. Science

2015;350:1199 – 204 .
123. Bollati V, Schwartz J, Wright R, et al. Decline in genomic DNA
139. Fadini GP, Losordo D, Dimmeler S. Critical reevaluation of endothelial
methylation through aging in a cohort of elderly subjects. Mech Ageing Dev
152. Sürder D, Manka R, Moccetti T, et al. Effect of bone marrow-derived
progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res
2009;130:234 – 9 .
mononuclear cell treatment, early or late after acute myocardial infarction:
2012;110:624 – 37 .
twelve months CMR and long-term clinical results. Circ Res 2016;119:481 – 90

124. Wang JC, Bennett M. Aging and atherosclerosis: mechanisms, .


140. Williamson K, Stringer SE, Alexander MY. Endothelial progenitor cells
functional consequences, and potential therapeutics for cellular
enter the aging arena. Front Physiol 2012;3:30 .
senescence. Circ Res 2012;111:245 – 59 .
153. Khan AR, Farid TA, Pathan A, et al. Impact of cell therapy on

myocardial perfusion and cardiovascular outcomes in patients with angina


141. Kushner EJ, MacEneaney OJ, Weil BR, et al. Aging is associated with
125. Illi B, Ciarapica R, Capogrossi MC. Chromatin methylation and refractory to medical therapy: a systematic review and meta-analysis. Circ
a proapoptotic endothelial progenitor cell phenotype. J Vasc Res 2011;48:
cardiovascular aging. J Mol Cell Cardiol 2015;83:21 – 31 . Res 2016;118:984 – 93 .
408 – 14 .

126. López-Otín C, Blasco MA, Partridge L, et al. The hallmarks of aging. 154. Rapola JM, Virtamo J, Ripatti S, et al. Randomised trial of a- tocopherol
142. Murasawa S, Llevadot J, Silver M, et al. Constitutive human
Cell 2013;153:1194 – 217 . and b- carotene supplements on incidence of major coronary events in men
telomerase reverse transcriptase expression enhances regenerative
with previous myocardial infarction. Lancet 1997;349:1715 – 20 .
127. Brunet A, Berger SL. Epigenetics of aging and aging-related disease. J properties of endothelial progenitor cells. Circulation 2002; 106:1133 – 9 .
Gerontol A Biol Sci Med Sci 2014;69 Suppl 1:S17 – 20 .

155. Jarski RW, Hightower KR, Dangovian MI. Vitamin E supplementation,


128. Hasegawa Y, Saito T, Ogihara T, et al. Blockade of the nuclear factor- k 143. He T, Joyner MJ, Katusic ZS. Aging decreases expression and activity
cardiovascular events, and cancer. JAMA 2005;294:425 – 6; author reply 426 .
B pathway in the endothelium prevents insulin resistance and prolongs life of glutathione peroxidase1 in human endothelial progenitor cells. Microvasc
spans. Circulation 2012;125:1122 – 33 . Res 2009;78:447 – 52 .

156. Coletta AP, Clark AL, Banarjee P, et al. Clinical trials update:
129. Schones DE, Leung A, Natarajan R. Chromatin modi fi cations 144. Endtmann C, Ebrahimian T, Czech T, et al. Angiotensin II impairs
RENEWAL (RENAISSANCE and RECOVER) and ATTACH. Eur J Heart
associatedwith diabetes and obesity. Arterioscler Thromb Vasc Biol endothelial progenitor cell number and function in vitro and in vivo:
Fail 2002;4: 559 – 61 .
2015;35:1557 – 61 . implications for vascular regeneration. Hypertension 2011;58:394 – 403 .

130. Paneni F, Volpe M, Lüscher TF, et al. SIRT1, p66Shc, and Set7/9 in
157. Venkatasubramanian S, Noh RM, Daga S, et al. Cardiovascular effects
vascular hyperglycemic memory: bringing all the strands together. Diabetes
of a novel SIRT1 activator, SRT2104, in otherwise healthy cigarette
2013;62:1800 – 7 . 145. Krankel N, Luscher TF, Landmesser U. Novel insights into vascular
smokers. J Am Heart Assoc 2013;2:e000042 .
repair mechanisms. Curr Pharm Des 2014;20:2430 – 8 .

131. Paneni F, Costantino S, Battista R, et al. Adverse epigenetic


158. Ungvari Z, Parrado-Fernandez C, Csiszar A, et al. Mechanisms
signatures by histone methyltransferase Set7 contribute to vascular 146. Paneni F, Costantino S, Kränkel N, et al. Reprogramming ageing and
underlying caloric restriction and lifespan regulation: implications for
dysfunction in patients with type 2 diabetes mellitus. Circ Cardiovasc Genet longevity genes restores paracrine angiogenic properties of early outgrowth
vascular aging. Circ Res 2008;102:519 – 28 .
2015;8:150 – 8 . cells. Eur Heart J 2016;37:1733 – 7 .

132. Xu S, Yin M, Koroleva M, et al. SIRT6 protects against endothelial 147. Mocharla P, Briand S, Giannotti G, et al. AngiomiR-126 expression
KEY WORDS arterial stiffness, endothelium, epigenetics,
dysfunction and atherosclerosis in mice. Aging (Albany NY) 2016;8:1064 – 82 and secretion from circulating CD34 þ and CD14 þ PBMCs: role for
stem cells
.

Anda mungkin juga menyukai