The placenta is a temporary organ that develops in most mammals during gestation, connecting the developing fetus to the uterine wall to facilitate the exchange of oxygen, nutrients, and waste products between the maternal and fetal circulations. In humans, it is a disc-shaped organ that develops in the uterus during pregnancy, connecting the fetus to the uterine wall through the umbilical cord.[1][2] It forms from fetal chorionic tissue and maternal endometrial tissue, known as the decidua, with implantation of the blastocyst occurring 6 to 10 days after fertilization.[2] By the end of the first trimester, the placenta is fully functional. It grows to resemble a spongy disc approximately 20 cm in diameter and 3 cm thick at term, composed of chorionic villi that project into maternal blood-filled spaces called the intervillous space.[2][3][4][5]In humans, the placenta's structure ensures efficient maternal-fetal exchange without direct mixing of blood, as fetal blood circulates within the villi while maternal blood bathes their exterior.[3] Key components include the umbilical cord, which contains two arteries and one vein for bidirectional flow, and the amniochorionic membrane that encloses the fetus.[2] It typically attaches to the upper or side wall of the uterus, though variations like placenta previa can occur when it lies low near the cervix.[1] The organ grows alongside the uterus throughout gestation, supporting fetal development until delivery, after which it is expelled as the afterbirth.[1][2]Physiologically, in humans, the placenta acts as the fetus's lifeline, facilitating gas exchange where oxygen diffuses from maternal blood to fetal blood and carbon dioxide moves in the opposite direction, primarily through passive diffusion across the thin placental barrier.[3] It transports essential nutrients like glucose, amino acids, and fatty acids via both passive and active mechanisms, while removing fetal waste products such as urea and bilirubin.[2] Additionally, the placenta serves endocrine functions by producing hormones including human chorionic gonadotropin (hCG) to maintain early pregnancy, progesterone to support uterine growth, and human placental lactogen (hPL) to enhance maternal glucose availability for the fetus.[3] It also provides a protective barrier, with syncytiotrophoblast cells and maternal macrophages shielding the fetus from pathogens and harmful substances.[3]
Evolutionary Aspects
Evolutionary Origins
The placenta is a temporary organ that develops during pregnancy in viviparous vertebrates, enabling the exchange of oxygen, nutrients, carbon dioxide, and waste products between the maternal and fetal circulations without direct blood mixing.[6] This structure has evolved independently at least 100 times across vertebrate lineages, including fish, amphibians, reptiles, and mammals, highlighting its recurrent adaptive value in supporting internal embryonic development.[7]Fossil evidence indicates that the earliest known instance of viviparity with inferred placentation dates to approximately 380 million years ago in the Devonian period, based on a placoderm fishfossil (Materpiscis attenboroughi) preserving an embryo connected by an umbilical cord, suggesting nutrient transfer mechanisms.[8] In amniote lineages, which originated around 310 million years ago during the Carboniferous period, viviparity and associated placental structures emerged as reptiles transitioned from oviparity, with direct fossil support appearing in Triassic marine reptiles such as ichthyosaurs around 250 million years ago, where embryos indicate internal gestation.[9] These origins reflect a gradual shift from egg-laying ancestors, driven by environmental pressures in aquatic or terrestrial habitats where external egg development was risky.Key evolutionary innovations include the fusion of extraembryonic membranes, such as the chorion and allantois, to form the chorioallantoic membrane in amniotes, which vascularizes and interfaces with uterine tissues to enhance gas and nutrient exchange.[7] In mammalian lineages, hemochorial placentation further advanced this by allowing direct contact between fetal trophoblast and maternal blood, a trait reconstructed as ancestral to eutherians around 100 million years ago.[10] Compared to oviparity, placentation provides adaptive advantages such as protection of embryos from predators, desiccation, and temperature fluctuations, along with sustained maternal provisioning that boosts offspring size and survival rates in unpredictable environments.[6]
Phylogenetic Diversity
The placenta exhibits remarkable phylogenetic diversity across vertebrates, with distinct types classified primarily by the degree of intimacy between maternal and fetal tissues. In mammals, placentas are categorized into three main types based on the layers separating maternal and fetal blood: epitheliochorial, where the uterine epithelium remains intact and multiple layers intervene (e.g., in horses and pigs); endotheliochorial, where trophoblast cells reach but do not breach maternal capillaryendothelium (e.g., in cats and dogs); and hemochorial, featuring direct contact between fetal trophoblast and maternal blood (e.g., in humans and rodents).[11][6] This classification, originally proposed by Grosser in 1909 and refined through comparative anatomy, reflects varying degrees of trophoblast invasiveness, with epitheliochorial being the least invasive and hemochorial the most.[12]Placental structures are predominantly found in mammals, particularly eutherians, where they evolved as the primary interface for nutrient and gas exchange, though marsupials possess a transient, yolk-sac-based epitheliochorial placenta during their short gestation.[6] Beyond mammals, viviparous reptiles such as squamate lizards (e.g., in the genus Mabuya) exhibit epitheliochorial or regionally invasive placentas that support extended embryonic development, representing convergent evolution independent of mammalian lineages.[6] Rare analogs occur in other vertebrates, including chondrichthyan fish like stingrays with trophonemata for nutrient provision and certain amphibians such as marsupial frogs (Gastrotheca), which utilize vascularized oviductal tissues for viviparity.[6] Phylogenetic analyses indicate that hemochorial placentation likely characterized the last common ancestor of eutherians around 100 million years ago, with subsequent transitions to less invasive forms in specific clades like afrotherians (epitheliochorial) and carnivorans (endotheliochorial).[13]Within eutherian mammals, placental diversity extends to macroscopic shapes, which correlate loosely with phylogeny and type: diffuse forms cover the entire chorion (e.g., in suids like pigs and equids like horses, epitheliochorial); zonary shapes form a belt around the fetus (e.g., in carnivores like dogs, endotheliochorial); cotyledonary types consist of multiple discrete attachment sites (e.g., in ruminants like cows, epitheliochorial); and discoid shapes appear as compact discs (e.g., in primates like humans and rodents, hemochorial).[11][13] These variations are distributed across the placental mammal phylogeny, with discoid and labyrinthine structures ancestral, while zonary and diffuse forms arose through independent evolutionary shifts in ungulates and carnivores, enhancing adaptability to diverse reproductive strategies.[13]Evolutionary trade-offs underpin this diversity, particularly in hemochorial placentas, where deep trophoblast invasion facilitates efficient nutrient transfer but heightens risks of maternal tissue damage, immune rejection, and pathogen transmission to the fetus.[6] To counter these risks, hemochorial systems have evolved specialized immune adaptations, such as regulatory T cells that suppress maternal anti-fetal responses and decidualization of the uterine lining to limit excessive invasion.[6] In contrast, epitheliochorial types minimize such risks through superficial attachment but may compromise exchange efficiency, illustrating a balance between fetal demands and maternal protection across phylogenetic lineages.[6]
Structure
Gross Anatomy
The human placenta is a disc-shaped organ that attaches to the uterine wall, measuring approximately 15-25 cm in diameter and 2-3 cm in thickness at term.[2][14] Its weight progresses from about 100 grams in the early second trimester to 500-600 grams at full term, representing roughly one-sixth of the fetal birth weight.[14][15] The spongy, discoid structure facilitates efficient exchange between maternal and fetal circulations, with the overall shape varying slightly but predominantly round or oval in uncomplicated pregnancies.[2][14]The fetal surface, known as the chorionic plate, appears smooth and shiny, covered by the amniochorionic membrane through which fetal blood vessels radiate from the umbilical cord insertion site.[2] The umbilical cord typically attaches centrally to this plate, though eccentric or marginal insertions occur in about 70-80% of cases.[14] On the opposing maternal surface, the basal plate is rough and divided into 15-28 cotyledons—convex, polygonal compartments formed by decidual septa that compartmentalize the intervillous space.[2][16][17]The amniotic membrane, the innermost layer, directly contacts the fetus and forms the amniotic sac filled with fluid, while the outer chorionic membrane fuses with the decidua to anchor the placenta and encloses the entire gestational sac, providing barrier protection and structural support.[2]Placental variations include the bilobate form, where the disc divides into two nearly equal lobes connected by membranes, occurring in 2-8% of pregnancies and potentially increasing risks of bleeding or retained tissue.[18] Succenturiate placentas feature one or more accessory lobes separate from the main disc but connected by vessels, with an incidence of 0.6-1.9%, raising concerns for vasa previa or postpartum hemorrhage if undiagnosed.[19][18] These macroscopic anomalies, while uncommon, highlight the placenta's adaptability compared to more uniform shapes in other mammals.[20]
Histology and Cellular Composition
The human placenta exhibits a hemochorial structure, characterized by maternal blood directly bathing the trophoblast layers without an intervening maternal epithelial barrier.[21] This microscopic organization centers on chorionic villi projecting into intervillous spaces filled with maternal blood, facilitating exchange while maintaining separation via fetal-derived layers. The primary barrier consists of the syncytiotrophoblast as the outermost multinucleated layer, underlain by the cytotrophoblast, with the villous core containing mesenchymal stroma and fetal capillaries lined by endothelium.[22][23]Key cellular components include the syncytiotrophoblast (STB), a continuous, non-proliferative sheet formed by fusion of cytotrophoblast cells, which covers the villous surface and features microvilli for enhanced surface area.[23] Beneath it lies the cytotrophoblast (CTB), a discontinuous monolayer of mononuclear progenitor cells that proliferate and differentiate into STB or extravillous trophoblasts, becoming sparse (about 25% coverage) by term.[22]Hofbauer cells, fetal-derived macrophages, reside in the villous stroma, appearing around day 18 post-fertilization and contributing to immune modulation and tissue remodeling.[24]Extravillous trophoblasts (EVTs) originate from CTB columns at anchoring villi, migrating into the decidua as interstitial or endovascular subtypes to remodel maternal spiral arteries.[23]Villous architecture evolves from primary villi (trophoblast only, around day 13) to secondary villi (with mesenchymal cores by day 16-17) and tertiary villi (incorporating fetal vessels by week 5), forming branching trees immersed in intervillous spaces.[22] These terminal villi, predominant by the third trimester, measure about 70-100 µm in diameter, with dense stroma enclosing 3-5 capillaries and occasional syncytial knots—protrusions of hyperchromatic STB nuclei indicating turnover.[24]Histological features change progressively across gestation. In the first trimester, villi are elongated with prominent CTB layers and nucleated fetal red blood cells; Hofbauer cells are sparse, and EVTs actively invade.[24] By the second trimester, intermediate villi develop loose stroma rich in Hofbauer cells, with emerging terminal villi and initial syncytial knots.[23] In the third trimester, CTB coverage thins, vasculosyncytial membranes form for closer capillary apposition to STB, and syncytial knots increase to about 30% of villi, reflecting accelerated STB renewal.[24] Standard histological assessment employs hematoxylin and eosin staining to visualize these layers, with immunohistochemistry (e.g., for hCG) highlighting STB and CD68 for Hofbauer cells.[24]
Gene and Protein Expression
The placenta exhibits a unique genomic profile characterized by the expression of specific genes that support its specialized functions. The alpha subunit of human chorionic gonadotropin (hCG) is encoded by the CGA gene on chromosome 6q14-q21, which is highly expressed in syncytiotrophoblast cells and shared among glycoprotein hormones.[25] The beta subunits of hCG are encoded by a cluster of genes (CGB) on chromosome 19q13.3, with CGB8, CGB5, and CGB7 showing predominant expression in first-trimester trophoblast, where CGB8 contributes the most to hCG production in both normal and pathological placentas.[26]Key protein markers in placental tissues include human placental lactogen (hPL), encoded by the CSH1 gene within the growth hormone/placental lactogen (GH/PL) cluster on chromosome 17q23, which is abundantly synthesized in the syncytiotrophoblast and exhibits elevated expression levels (up to 9,508 nTPM) specific to this tissue.[27] Syncytin, derived from the human endogenous retrovirus envelope gene ERVW-1 (also known as HERV-W-Env), promotes trophoblast cell fusion essential for syncytiotrophoblast formation and is highly enriched in placental tissue (148 nTPM), with expression primarily in multinucleated syncytia.[28]Epigenetic regulation plays a critical role in placental gene expression, particularly through genomic imprinting, where approximately 100 imprinted genes are expressed in the human placenta, many of which are essential for trophoblast proliferation and differentiation, such as PEG10 and PHLDA2.[29] In trophoblast cells, X-chromosome inactivation (XCI) is imprinted, preferentially silencing the paternal X chromosome in extraembryonic lineages via mechanisms involving the non-coding RNA Xist, which helps maintain dosage compensation and placental stability.[29][30]Transcriptomic analyses reveal a distinct placental gene expression landscape, with approximately 300 genes showing elevated expression compared to other tissues, including around 70 tissue-enriched protein-coding genes involved in processes like trophoblastinvasion and angiogenesis.[31] For instance, genes such as MMP12 (upregulated 186-fold in early gestation) facilitate extracellular matrix remodeling for invasion, while ANGPT4 (218-fold upregulation) supports vascular remodeling in angiogenesis.[32] These placenta-enriched genes, identified through RNA-seq comparisons with multi-tissue datasets like GTEx, underscore the organ's molecular specialization.[33]
Development
Early Formation and Implantation
The formation of the placenta begins shortly after fertilization with the development of the zygote into a multicellular embryo. Following fertilization, the zygote undergoes rapid cleavage divisions, progressing from a 2-cell stage on day 1 to a morula of approximately 16 cells by day 3-4 post-fertilization.[34] By day 5, the embryo transforms into a blastocyst through cavitation, forming a fluid-filled blastocoel cavity enclosed by the trophectoderm (TE), an outer epithelial layer that surrounds the inner cell mass (ICM).[34] The TEdifferentiation is driven by transcription factors such as CDX2, which specify the outer cells for placental lineage while the ICM commits to embryonic tissues.[35]Implantation marks the critical transition from free-floating blastocyst to uterine attachment, occurring between days 6 and 10 post-fertilization in humans. This process unfolds in sequential phases: apposition, where the polar TE of the blastocyst loosely contacts the receptive endometrial epithelium around day 6; adhesion, which strengthens this contact by day 7 through molecular interactions like integrins and selectins; and invasion, where trophoblast cells penetrate the endometrial stroma by days 8-10, embedding the blastocyst within the decidua.[36] The uterine environment must be receptive, influenced by the post-ovulatory progesterone surge that induces endometrial changes, including spiral artery dilation and immune modulation, to facilitate this attachment without rejection.As implantation proceeds, the TE undergoes differentiation into distinct trophoblast subtypes essential for placental establishment. The TE first forms a monolayer that rapidly differentiates into cytotrophoblast (CTB), a proliferative stem-like layer of mononucleated cells, and syncytiotrophoblast (STB), a multinucleated, invasive layer generated by CTB fusion. The STB leads the initial invasion, secreting proteases to breach the endometrial basement membrane, while CTB provides progenitors for further placental structures. This differentiation is regulated by pathways involving GATA3 and TFAP2C, ensuring balanced proliferation and fusion.[37]Hormonal signals orchestrate these early events, with progesterone playing a pivotal role in endometrial preparation and trophoblast function. Progesterone, secreted by the ovarian corpus luteum, promotes uterine receptivity by upregulating genes for adhesion molecules and suppressing inflammatory responses, creating a window of implantation typically 6-10 days after ovulation. Concurrently, the emerging STB produces human chorionic gonadotropin (hCG), detectable in maternal serum by day 8-9, which sustains corpus luteum function to prolong progesterone secretion and prevent menstruation. hCG also directly enhances trophoblast invasion and immune tolerance at the maternal-fetal interface.
Maturation and Growth Stages
Following implantation, the placenta undergoes progressive morphological and functional maturation to support escalating fetal demands. In the first trimester, primary chorionic villi emerge as finger-like projections from the trophoblast layer around weeks 2-3 post-fertilization, evolving into secondary villi with mesenchymal cores by week 3 and tertiary villi with fetal blood vessels by week 4, establishing the foundational structure for nutrient exchange.[2] This villous formation phase is characterized by rapid proliferation of cytotrophoblasts and syncytiotrophoblasts, increasing the placental surface area to accommodate initial embryonic growth.[38]During the second trimester, placental growth accelerates with peak vascularization, as fetal capillaries expand within the villi and maternal spiral arteries remodel to enhance blood flow into the intervillous space around weeks 10-12.[2] Branching of villi intensifies, forming complex trees that boost the exchange surface area by up to 10-15 fold compared to early gestation, adapting to rising oxygen and nutrient requirements for fetal organogenesis.[38] Placental weight rises exponentially during this period, from approximately 50 g at 10-12 weeks to around 200 g by week 20, reflecting heightened metabolic activity and structural expansion.[39]In the third trimester, the placenta reaches full maturation, with villi differentiating into terminal types optimized for diffusion and early signs of senescence such as calcification appearing in the basal plate and villous cores from week 28 onward, indicating adaptive aging to prioritize fetal viability.[2] Weight continues to increase to about 500-600 g at term, though growth rate slows as the organ prioritizes efficiency over further expansion.[38] This phase features further villous branching and syncytial knot formation, enhancing gas exchange capacity amid peak fetal demands.[40]Recent studies using single-cell transcriptomics have elucidated dynamic trophoblast differentiation pathways influenced by oxygen gradients and maternal metabolites, providing insights into placental adaptability and potential links to disorders like preeclampsia (as of 2023).[41]Placental maturation is influenced by environmental factors, including oxygen tension, which remains low (~20 mmHg or 2.5% O₂) in the first trimester to promote trophoblastproliferation but rises to ~60 mmHg by week 14, driving vascular remodeling and differentiation.[42] Maternal nutrition also plays a key role, with adequate intake of glucose, amino acids, and lipids supporting transporter expression (e.g., GLUT1 for glucose) and villous development; deficiencies can impair growth and lead to restricted surface area expansion.[43] These adaptations ensure the placenta's surface area expands from ~4 m² in mid-gestation to ~14 m² at term via iterative villous branching, optimizing transfer efficiency without proportional weight gain.[2]
Placental Circulation
Maternal Circulation
The maternal circulation to the placenta is primarily supplied by the endometrial spiral arteries, which undergo extensive remodeling to accommodate the increased demands of pregnancy. These arteries, originating from the uterine radial arteries, deliver oxygenated blood directly into the intervillous spaces of the placenta. At term, uteroplacental blood flow reaches approximately 600-700 mL/min, accounting for about 80% of total uterine perfusion and ensuring adequate nutrient delivery to the fetus.[44]A critical adaptation involves the invasion of extravillous trophoblast cells, which penetrate the decidua and myometrium to remodel the spiral arteries into wide, low-resistance conduits. This process replaces the muscular and endothelial linings of the arteries with fibrinoid material and trophoblast cells, reducing vascular resistance and preventing vasospasm. The remodeling begins in the first trimester and is essential for progressively increasing blood flow as gestation advances.[45][46]Maternal blood enters the intervillous spaces through the dilated orifices of the spiral arteries in a high-velocity, jet-like manner, typically at speeds of 1-2 m/s, before slowing and pooling around the fetal villi. This pulsatile inflow creates a dynamic mixing environment within the intervillous spaces, with blood draining subsequently through endometrial veins back into the maternal systemic circulation. The overall flow is non-laminar and influenced by uterine contractions, optimizing exposure to the placental exchange surface.[47][48]Pregnancy induces systemic hypervolemia, expanding maternal blood volume by 40-50% through increased plasma volume and red cell mass, which supports the elevated uteroplacental perfusion requirements. This physiological change peaks in the third trimester and contributes to the overall cardiovascular adaptations necessary for placental function.[49]
Fetoplacental Circulation
The fetoplacental circulation encompasses the vascular network on the fetal side of the placenta, facilitating the transport of oxygenated blood and nutrients from the placenta to the fetus while returning deoxygenated blood and waste products. This system begins with the umbilical cord, which contains two umbilical arteries carrying deoxygenated fetal blood from the internal iliac arteries toward the placenta and a single umbilical vein returning oxygenated blood to the fetus. These vessels are embedded in Wharton's jelly, a mucoid connective tissue that provides cushioning, structural support, and protection against compression and mechanical stress during fetal movement.[50][51]From the umbilical arteries, blood branches into chorionic arteries on the fetal surface of the placenta, further dividing into smaller vessels that form extensive capillary beds within the chorionic villi. These capillary networks, surrounded by the villous trophoblast, enable efficient crosscurrent exchange of gases, nutrients, and waste between fetal blood and maternal blood in the adjacent intervillous spaces, without direct mixing of the circulations. The arrangement maximizes diffusion gradients across the thin placental barrier, supporting fetal oxygenation and nutrition.[44][52]Blood flow dynamics in the fetoplacental circulation are characterized by a low-resistance pathway, with total flow through the umbilical vein reaching approximately 350–400 mL/min at term, representing about 40% of the fetal cardiac output. Upon entering the fetus via the umbilical vein, approximately 20-30% of the blood is shunted through the ductus venosus directly to the inferior vena cava, bypassing the liver to prioritize vital organ perfusion.[53] The fetal heart adaptations, including a right ventricular dominance with higher pressures on the right side (approximately 50–60 mmHg in the pulmonary artery and descending aorta), generate pressure gradients that maintain unidirectional flow: fetal arterial pressure exceeds the maternal intervillous space pressure (about 10 mmHg), while venous pressure remains low (around 5-10 mmHg), preventing backflow and ensuring efficient circulation complementary to the maternal uterine blood supply.[54]
Physiological Functions
Nutrient and Gas Exchange
The placenta facilitates the exchange of oxygen and carbon dioxide between maternal and fetal blood primarily through passive diffusion across the syncytiotrophoblast layer, driven by partial pressure gradients. In late gestation, the partial pressure of oxygen (PO₂) in maternal uterine venous blood is approximately 40-50 mmHg, while in fetal umbilical venous blood it is about 20-30 mmHg, creating a favorable gradient for oxygen to diffuse from the maternal intervillous space into the fetal capillaries.[55] This process is enhanced by the higher oxygen affinity of fetal hemoglobin compared to maternal hemoglobin, which allows fetal blood to extract oxygen more efficiently at lower partial pressures, with fetal hemoglobin's P50 value around 19 mmHg versus 27 mmHg for adult hemoglobin.[56][57] Carbon dioxide removal occurs similarly via diffusion, as fetal blood has a higher PCO₂ (about 46-50 mmHg) than maternal blood (about 40 mmHg), promoting transfer from fetus to mother; this is facilitated by the Haldane effect, where deoxygenation of maternal blood increases its CO₂ carrying capacity.[58]The rate of gas diffusion across the placental barrier follows Fick's first law, which states that the flux of a gas is proportional to the surface area available for diffusion, the diffusion coefficient of the gas, and the partial pressure difference, divided by the thickness of the barrier. At term, the placental villous surface area reaches approximately 12-14 m², optimizing exchange capacity, while the barrier thickness thins to 2-4 μm, minimizing diffusion distance and enhancing efficiency.[59][60] These structural adaptations, combined with differences in hemoglobin oxygen affinity, ensure that the placenta can meet the fetus's oxygen demands, which increase from about 3-5 mL/min/kg in early pregnancy to approximately 6-8 mL/min/kg at term, without direct mixing of maternal and fetal blood streams.[61][62]Nutrient transfer, particularly of glucose and amino acids, involves facilitated and active transport mechanisms to support fetal growth against concentration gradients. Glucose, the primary energy substrate, crosses the placenta via facilitated diffusion mediated by glucose transporter proteins, predominantly GLUT1 on the maternal-facing basal membrane and GLUT3 on the fetal-facing microvillous membrane of the syncytiotrophoblast, enabling net transfer despite similar maternal and fetal plasma concentrations due to fetal utilization.[63][64] Amino acids are transported actively via sodium-dependent systems on the microvillous membrane (e.g., System A for small neutral amino acids) and sodium-independent exchangers like System L (involving LAT1 and LAT2 isoforms) on the basal membrane, maintaining higher fetal plasma levels (up to 2-3 times maternal concentrations for essential amino acids like leucine) to fuel protein synthesis.[65][66] The expansive villous surface area and thin barrier further amplify these transport rates, ensuring adequate nutrient delivery without compromising the barrier's integrity.[67]
Waste Excretion and Metabolism
The placenta plays a crucial role in fetal waste excretion by facilitating the diffusion of metabolic byproducts from the fetal circulation to the maternal bloodstream, preventing their accumulation in the fetus. Urea and creatinine, primary nitrogenous wastes from fetal protein metabolism, cross the placental barrier via passive diffusion driven by concentration gradients across the syncytiotrophoblast layer. This process ensures efficient clearance without direct mixing of maternal and fetal blood, relying on the thin placental membrane for selective permeability. Similarly, unconjugated bilirubin, a byproduct of fetal heme breakdown, diffuses from fetal to maternal blood, where it is processed by the maternal liver for conjugation and excretion.[2][68]In addition to waste removal, the placenta actively participates in metabolic transformations to support fetal needs and detoxification. It metabolizes maternal lipids by taking up lipoproteins through receptor-mediated endocytosis, followed by hydrolysis via enzymes such as lipoprotein lipase and phospholipases, releasing free fatty acids in forms more readily utilizable by the fetus for membranesynthesis and energy. This conversion process adapts maternal triacylglycerols and cholesterol into bioavailable components, with fetal biosynthesis supplementing supply in later gestation. For detoxification, placental cytochrome P450 enzymes, including CYP1A1, CYP2E1, and CYP3A isoforms, oxidize xenobiotics and endogenous toxins, reducing their potential harm to the fetus through phase I metabolism. These enzymes exhibit gestational variation in expression and activity, enhancing the placental barrier against environmental insults.[69][70]The placenta's own metabolic demands are substantial, consuming approximately 30% of the maternal glucose supply to generate ATP via glycolysis and oxidative phosphorylation, thereby sustaining its transport and synthetic functions. This high energy requirement underscores the placenta's role as a semi-autonomous organ, prioritizing its viability to maintain fetal homeostasis. Effective bilirubin handling by the placenta and maternal liver is particularly vital, as it averts fetal hyperbilirubinemia and the risk of kernicterus—a neurotoxic condition from unbound bilirubin crossing the blood-brain barrier—ensuring neurological protection during development.[71][68]
Endocrine and Hormonal Regulation
The placenta functions as a temporary endocrine organ, producing a suite of hormones essential for maintaining pregnancy, supporting fetal development, and preparing for parturition. These hormones are primarily synthesized by the syncytiotrophoblast layer, with some contributions from cytotrophoblasts and other placental cells. Key among them are human chorionic gonadotropin (hCG), progesterone, estrogens (particularly estriol), relaxin, and corticotropin-releasing hormone (CRH), each playing distinct roles in maternal and fetal physiology through complex regulatory mechanisms.[72]Human chorionic gonadotropin (hCG), secreted by syncytiotrophoblasts shortly after implantation, is crucial for early pregnancy maintenance by stimulating the corpus luteum to produce progesterone until the placenta assumes this role around 7-10 weeks. hCG levels peak at approximately 100,000-110,000 mIU/mL between 8-10 weeks of gestation, then decline to about 10% of peak by 22 weeks before a gradual rise toward term. This hormone also exhibits mild thyrotropic and immunosuppressive effects, aiding maternal adaptation.[73][72]Progesterone, produced by the syncytiotrophoblast from maternal cholesterol precursors, is vital for uterine quiescence, preventing contractions and supporting decidualization to sustain implantation. Its levels rise progressively, reaching 100-200 ng/mL by term, with the placenta synthesizing about 250 mg daily. This hormone inhibits myometrial activity and modulates immune responses, ensuring a stable intrauterine environment.[73][72]Estrogens, including 17β-estradiol and estriol, are generated through the feto-placental unit, where the placenta aromatizes fetal adrenal precursors like dehydroepiandrosterone sulfate (DHEAS). Estriol, the predominant form, promotes cervical ripening and increases uteroplacental blood flow, while overall estrogen levels (6-30 ng/mL for 17β-estradiol and 10-30 ng/mL for estriol at term) support mammary gland development and fetal maturation. These hormones also influence progesterone metabolism, contributing to the shift toward labor.[72]As pregnancy advances, relaxin and CRH emerge in roles tied to labor initiation. Relaxin, secreted by the placenta and fetal membranes, softens the cervix and relaxes pelvic ligaments, facilitating delivery; its concentrations increase in the first trimester and peak near term. Placental CRH, produced by cytotrophoblasts, stimulates the fetal pituitary-adrenal axis by promoting adrenocorticotropic hormone (ACTH) release, which in turn elevates cortisol levels in a positive feedback loop that augments CRH production and triggers prostaglandin synthesis for uterine contractions. CRH levels remain low in early pregnancy but rise dramatically in the third trimester, peaking at delivery.[72]
Immune Protection and Tolerance
The placenta establishes a critical immunological barrier to protect the semi-allogeneic fetus from maternal immune rejection while promoting tolerance at the maternal-fetal interface. This dual function is essential for successful pregnancy, as the fetus expresses paternal antigens that could otherwise trigger an immune response. The trophoblast cells, which form the outer layer of the placenta, play a central role in this process by limiting immune cell access and modulating maternal immunity.[74]A key component of the immunological barrier is the expression of human leukocyte antigen-G (HLA-G) by extravillous trophoblast cells, which inhibits the cytotoxic activity of maternal natural killer (NK) cells and T-cells. HLA-G binds to inhibitory receptors such as KIR2DL4 on NK cells and ILT2/ILT4 on T-cells, delivering signals that suppress their activation and prevent lysis of trophoblast cells. This expression is tightly regulated by a trophoblast-specific enhancer, ensuring HLA-G is predominantly active at the placental interface to maintain immune privilege without compromising overall maternal immunity.[75][76]Tolerance mechanisms further support this protection through the induction of regulatory T-cells (Tregs) and a shift in cytokine profiles toward an anti-inflammatory state. During early pregnancy, exposure to paternal antigens via seminal fluid and placental signals expands the pool of CD4+CD25+Foxp3+ Tregs, which suppress effector T-cell responses and promote long-term tolerance to fetal antigens. Concurrently, the placenta environment favors a Th2 cytokine bias, with elevated levels of interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) that dampen pro-inflammatory Th1 responses and enhance Treg function. These cytokines are secreted by trophoblast cells and decidual immune cells, creating a localized immunosuppressive milieu that sustains gestation.[77][78][79]Epigenetic modifications, particularly DNA methylation, contribute to immune gene silencing in trophoblast cells, reinforcing tolerance by repressing expression of classical HLA class I and II genes that could provoke maternal T-cell recognition. In trophoblast lineages, hypermethylation of promoters for genes like HLA-A, HLA-B, and CIITA (a master regulator of MHC II) occurs during differentiation, ensuring these immunogenic molecules remain transcriptionally inactive. This trophoblast-specific epigenome is established early in development and maintained by DNA methyltransferases, providing a stable molecular basis for immune evasion.[80][81][82]Disruptions in these mechanisms, such as reduced HLA-G expression or impaired Treg induction, are associated with preeclampsia, where failed tolerance leads to excessive inflammation and placental dysfunction; however, in normal pregnancies, these processes robustly safeguard fetal development. Complementing these molecular strategies, the histological layers of the syncytiotrophoblast serve as a physical barrier that further restricts direct immune cell contact with fetal tissues.[83]
Microbiome Interactions
The existence of a distinct placental microbiome remains a topic of significant debate in scientific literature as of 2025. While some studies using 16S rRNA gene sequencing have reported a low-biomass community of microorganisms in placental tissue, typically on the order of 10^3 to 10^4 bacterial cells per gram, others attribute these findings to contamination during sampling, processing, or analysis rather than a true resident microbiota.[84][85] Reported microbial profiles, when present, are predominantly composed of nonpathogenic commensal bacteria from phyla such as Firmicutes and Proteobacteria, with genera including Lactobacillus and Bifidobacterium, resembling maternal vaginal or skin flora.[86] These populations, if real, would be sparse compared to microbiomes in other body sites, potentially reflecting the placenta's role in maintaining a relatively sterile environment during gestation.[87]Proposed origins of any placental microbiota include ascending migration from maternal vaginal and cervical flora or hematogenous seeding from the bloodstream, such as from oral or gut sources. Ascending pathways could allow vaginal microbes to reach the placenta during implantation, while hematogenous routes might enable transient bacterial translocation.[88][89] If a microbiome exists, it may modulate maternal-fetal immunity, nutrient metabolism, and barrier integrity, potentially influencing pregnancy outcomes by regulating inflammation or producing metabolites like short-chain fatty acids. Dysbiosis has been linked in some studies to risks such as preterm birth, though causality remains unestablished due to methodological concerns. Recent investigations continue to explore these associations, but consensus on the placenta's microbial status is lacking, with calls for improved controls to distinguish true signals from artifacts.[86][90][91]
Postpartum Processes
Separation and Delivery
The separation of the placenta from the uterine wall during the third stage of labor involves coordinated physiological and mechanical processes driven by myometrial contractions. Following fetal delivery, continued uterine contractions cause the myometrium to thicken and retract, reducing the surface area of the placental implantation site and generating shear forces between the elastic uterine wall and the relatively rigid placenta. These shear forces initiate detachment, typically starting at the periphery or a pole, while simultaneous compression of uterine veins leads to their rupture and the formation of a retroplacental hematoma—a collection of maternal blood that accumulates behind the placenta, further promoting separation by expanding the potential space between the placenta and decidua. This hematoma, combined with the shearing action, results in the placenta becoming partially or fully detached within 5-10 minutes in most cases, after which additional contractions expel it from the uterus.[92][93]The management of placental delivery falls into two primary approaches: active and expectant. Active management incorporates prophylactic uterotonics, such as oxytocin administered intramuscularly (10 IU) or intravenously (10-20 IU in 500-1000 mL fluid) immediately after birth, to augment myometrial contractions and accelerate separation. This approach shortens the third stage duration to typically less than 30 minutes in over 95% of cases, compared to 20-60 minutes with expectant management, which awaits natural signs of separation without pharmacological intervention. Expectant management promotes physiological expulsion through ongoing spontaneous contractions but carries a higher risk of prolonged third stage and postpartum hemorrhage.[94][95]To facilitate expulsion once separation occurs, controlled cord traction is often employed in active management protocols. This technique involves steady, gentle downward traction on the umbilical cord while a clinician applies counter-pressure to the lower uterine segment via bimanual compression to stabilize the uterus and prevent inversion or avulsion. Key clinical signs indicating separation include a sudden lengthening of the umbilical cord (as the placenta descends into the lower uterine segment), a gush of blood (approximately 100-250 mL from the retroplacental hematoma), and a rise in the uterine fundus with a globular, firmer contour upon abdominal palpation. These signs confirm readiness for traction, ensuring safe delivery without undue force.[93][95]Historically, placental expulsion relied heavily on manual removal under general anesthesia due to retained placenta risks, a practice common until the early 20th century. A pivotal shift began in the 1910s with the introduction of pharmacological aids; in 1912, Barry Hart advocated prophylactic intramuscular ergot administration after fetal delivery to prevent hemorrhage, marking the origins of active management. By the 1950s-1960s, ergot was largely replaced by synthetic oxytocin (Pitocin), which offered faster onset and fewer side effects, further diminishing the need for manual interventions and standardizing uterotonic use worldwide.[96][97]
Clinical Examination and Retention Risks
Following the separation and delivery of the placenta, a thorough clinical examination is essential to confirm its completeness and prevent complications. The gross inspection begins with assessing the overall shape and size of the placenta, which typically measures 15-20 cm in diameter and weighs 400-600 grams in a term pregnancy. The maternal surface is examined for the presence of all 15-30 cotyledons, ensuring no missing fragments that could indicate retention; the fetal surface is checked for intact vessels branching from the umbilical cord without disruptions suggesting avulsion. The membranes are inspected for completeness, with the amnion and chorion forming a continuous sac that inserts at the placental margin, and the rupture site measured to verify it aligns with the gestational age. This initial bedside evaluation by the obstetric provider helps triage for further pathologic analysis if abnormalities are noted.[98][99][100]Retained placenta is diagnosed when the placenta or its fragments fail to deliver spontaneously within 30 minutes after birth in active management of the third stage or 60 minutes in physiological management, occurring in approximately 2-3% of vaginal deliveries. A primary risk is postpartum hemorrhage (PPH), which arises from inadequate uterine contraction due to retained tissue and accounts for up to 20% of severe PPH cases, with blood loss exceeding 500 mL potentially leading to hypovolemic shock if untreated. Management typically involves manual removal of the placenta under regional or general anesthesia to ensure patient comfort and safety, as the procedure requires cervical dilatation and uterine exploration.[101][102][103]If gross inspection suggests remnants or symptoms like persistent bleeding occur, transvaginal ultrasound is the primary diagnostic tool, identifying echogenic masses or thickened endometrium (>10 mm) with a sensitivity of about 75% for retained products of conception. Treatment protocols emphasize prompt intervention: manual removal or dilatation and curettage should occur within 30 minutes of diagnosis to minimize hemorrhage risk and prevent maternal shock, often guided by ultrasound to target fragments precisely.[104][105][101]Long-term effects of retained placenta and its management include a heightened risk of endometritis or pelvic infection from bacterial ascension into the uterus, occurring in up to 15-20% of cases without timely removal. Aggressive curettage for remnants can lead to Asherman's syndrome, characterized by intrauterine adhesions that impair menstrual function and fertility in 1-5% of affected patients, potentially requiring hysteroscopic adhesiolysis for resolution.[106][107]
Clinical Significance
Placental Disorders and Complications
Placental disorders encompass a range of pathological conditions that impair the formation, attachment, or function of the placenta, leading to adverse maternal and fetal outcomes during pregnancy. These disruptions often stem from abnormalities in trophoblast invasion, vascular remodeling, or inflammatory processes, which compromise the placenta's role in nutrient and gas exchange. Common manifestations include maternal hypertension, bleeding, and fetal growth restriction, with severe cases necessitating preterm delivery or emergency interventions.[108]Preeclampsia is a multisystem disorder characterized by shallow placental implantation, where cytotrophoblast invasion into the uterine wall is insufficient, resulting in incomplete remodeling of spiral arteries and persistent high-resistance blood flow. This leads to placental ischemia and release of anti-angiogenic factors like soluble fms-like tyrosine kinase-1 (sFlt-1), contributing to endothelial dysfunction and maternal symptoms such as hypertension and proteinuria after 20 weeks of gestation.[109][110][111]Placental abruption involves premature separation of the normally attached placenta from the uterine wall, typically after 20 weeks of gestation, with an incidence of approximately 0.5-1% of pregnancies. This separation disrupts maternal-fetal blood flow, causing retroplacental hemorrhage and potential fetal hypoxia or demise if extensive.[112][113]The placenta accreta spectrum refers to abnormal placental attachment where trophoblasts invade beyond the decidua into the myometrium or beyond, classified as accreta (superficial myometrial attachment), increta (deeper invasion), or percreta (penetration through the uterine wall). This condition arises from defects in the endometrial-myometrial interface, often leading to massive postpartum hemorrhage due to failure of placental separation.[114][115][116]Placental infarction, representing ischemic necrosis of villous tissue, and calcification, involving calcium deposits in the villous basement membrane or intervillous space, are physiological signs of placental aging in term pregnancies but become pathological when excessive, particularly in intrauterine growth restriction (IUGR). In IUGR cases, infarcts and calcifications indicate chronic uteroplacental insufficiency, reducing functional placental mass and impairing fetal nutrient supply.[117][118][119]Key risk factors for these disorders include maternal hypertension, which promotes vascular damage and ischemia; cigarette smoking, which induces vasoconstriction and oxidative stress; and prior cesarean sections, which scar the endometrium and predispose to abnormal implantation. These factors collectively heighten the risk of fetal hypoxia, preterm birth, and long-term neurodevelopmental issues in offspring.[113][120][121]Post-2020 studies have highlighted the role of SARS-CoV-2 infection in placental pathology, with evidence of vertical transmission in rare cases via transplacental spread, often accompanied by syncytiotrophoblast infection and chronic villitis leading to inflammation and increased risk of fetal demise. These findings underscore SARS-CoV-2's potential to exacerbate placental insufficiency through direct viral effects and maternal inflammatory responses.[122][123][124]
Diagnostic Methods and Research Advances
Diagnostic methods for assessing placental health primarily rely on non-invasive imaging techniques and biomarkers, supplemented by invasive procedures when necessary. Doppler ultrasound is widely used to evaluate blood flow in the utero-placental circulation, measuring waveform indices and notching to identify pregnancies at risk for complications such as fetal growth restriction.[125] This technique provides real-time assessment of vascular resistance in the uterine arteries and umbilical cord, aiding in the early detection of impaired perfusion.[126]Magnetic resonance imaging (MRI), particularly blood oxygen level-dependent (BOLD) MRI, offers a non-invasive means to quantify placental oxygenation by detecting changes in deoxyhemoglobin levels during maternal oxygen challenges.[127] Advanced MRI protocols, including oxygen-enhanced imaging, have demonstrated the ability to differentiate normal from pathological placentas, such as those affected by insufficiency, with high sensitivity to oxygenation gradients.[128]Biomarkers in maternal serum play a crucial role in predicting placental dysfunction, with placental growth factor (PlGF) emerging as a key indicator for preeclampsia risk. Low PlGF levels in the second trimester correlate strongly with subsequent hypertensive disorders, offering predictive accuracy when combined with soluble fms-like tyrosine kinase-1 (sFlt-1) ratios, with sensitivities up to 93% in high-risk cohorts.[129] The sFlt-1/PlGF ratio, cleared by the FDA for clinical use, helps rule out preeclampsia within four weeks when below 38, guiding triage and monitoring.[130] For invasive diagnostics, chorionic villus sampling (CVS) involves extracting placental tissue between 10 and 13 weeks of gestation to analyze fetal DNA for chromosomal abnormalities and genetic disorders, providing definitive results with a detection rate exceeding 99% for conditions like Down syndrome.[131] Though carrying a small risk of miscarriage (about 0.5-1%), CVS remains essential for high-risk pregnancies identified by noninvasive prenatal testing.[132]Research advances in placental diagnostics and therapeutics have accelerated, particularly with stem cell applications and in vitro models. Placental-derived mesenchymal stem cells (PMSCs) have shown promise in regenerative therapies, with clinical trials since 2015 demonstrating safety in treating conditions like myelomeningocele through fetal interventions, where PMSC infusions improved ambulation in preclinical ovine models.[133] Preclinical studies and emerging clinical trials, including those using mesenchymal stem cell (MSC)-derived exosomes, explore their potential for vascular repair following preeclampsia, leveraging immunomodulatory and angiogenic properties without tumorigenic risks observed in other stem cell sources.[134][135] Placental organoid models, derived from trophoblast cells, replicate villous structures for high-throughput drug testing, enabling evaluation of transport across the syncytiotrophoblast barrier and teratogenic effects, as validated in 2023 protocols that mimic first-trimester physiology.[136] These 3D bioprinted organoids, advanced in 2025, facilitate screening for placental dysfunction therapies by simulating maternal-fetal exchange.[137]Emerging 2023-2025 developments highlight exosome-based diagnostics from placental sources, where circulating placental exosomes serve as non-invasive biomarkers for maternal diseases like preeclampsia, detectable via surface markers such as placental alkaline phosphatase.[138] Studies have identified exosomal microRNAs and proteins that predict complications with improved specificity over traditional markers, paving the way for liquid biopsy integration in routine screening.[139]Artificial intelligence enhances imaging analysis, with deep learning models achieving over 95% accuracy in segmenting placental ultrasound images to detect anomalies like accreta spectrum disorders.[140] AI-driven MRI radiomics, combining texture features with clinical data, predicts fetal growth restriction with AUC values above 0.85, supporting automated, explainable diagnostics in obstetrics.[141]
Societal and Historical Context
Cultural and Symbolic Roles
Across various indigenous cultures, the placenta holds profound ritual significance, often symbolizing a connection to ancestors and the land. In Māori tradition, the placenta, known as whenua, is buried in a sacred location to bind the child to their ancestral heritage and the earth, reinforcing familial and environmental ties.[142] Similarly, in Native Hawaiian practices, the placenta, or 'iēwe, is buried in the homeland to establish the child's spiritual link to place and forebears, with the umbilical cord (piko) sometimes placed in a ceremonial hole (puka) as part of this rite.[142] These burial rituals underscore the placenta's role as a twin or companion to the newborn, believed to influence the child's future well-being if not handled respectfully.[143]In some African and Asian postpartum traditions, the placenta is consumed for its purported vitality-enhancing properties. Among certain South African indigenous groups, portions of the placenta are dried, powdered, and ingested to promote fertility and health benefits postpartum.[144] In Traditional Chinese Medicine, dried placenta (ziheche) has been used for centuries by postpartum mothers to replenish energy, boost lactation, and restore vitality after childbirth.[145] These practices view the organ as a potent source of life force, though consumption remains selective and ritualistic rather than widespread.[145]Symbolically, the placenta has represented the source of life in artistic expressions, particularly during the Renaissance, when anatomical discoveries intertwined with spiritual themes. In Michelangelo's Creation of Adam fresco on the Sistine Chapel ceiling, the figures surrounding God have been interpreted as resembling a cross-section of a placenta and uterus, evoking themes of divine birth and human origination from a nurturing, life-sustaining organ.[146] This depiction highlights the placenta's metaphorical role as a bridge between creator and creation, blending emerging scientific understanding with religious symbolism.[146] In contrast, Western cultural attitudes historically treated the placenta as taboo, associating it with impurity and blood-related pollution, leading to its routine disposal as medical waste until the 20th century when biomedical views solidified its status as expendable tissue.[147]Modern trends have revived interest in the placenta's cultural roles, adapting ancient reverence to contemporary contexts. Lotus birth, a practice originating in the 1970s in California, involves leaving the umbilical cord uncut after delivery, allowing the placenta to remain attached to the newborn until it naturally detaches, typically within a week, to honor the organ's spiritual companionship.[148] Placenta encapsulation, a rising postpartum trend in industrialized nations, processes the dried placenta into capsules for maternal ingestion, claimed to support hormonal balance and energy, though rooted in selective interpretations of traditional uses.[149]Cross-culturally, the placenta's treatment varies starkly between veneration and utilitarian disposal. In Balinese Hinduism, the placenta undergoes ceremonial rites as part of sacred postpartum worship, symbolizing the soul's earthly imprint and communal harmony.[150] This contrasts with biomedical practices in modern healthcare systems, where the placenta is typically discarded as biohazardous waste post-delivery, prioritizing clinical efficiency over symbolic meaning.[142]
Historical and Medical Evolution
In ancient times, the placenta was often regarded as superfluous waste material following delivery. Aristotle (384–322 BCE), in his writings on reproduction, described the placenta as the "afterbirth" and recommended tying the umbilical cord away from it upon expulsion to avoid contamination, reflecting a view of it as discardable residue rather than a vital organ.[151] Similarly, Hippocratic theories (circa 460–370 BCE) posited that the fetus derived nutrition from maternal blood, influencing early views on reproduction and placental expulsion as a natural process managed primarily by midwives.[152] These perspectives, rooted in observational midwifery, influenced early medical practices across the Mediterranean, occasionally intersecting with cultural rituals that dictated afterbirth disposal.[153]A pivotal milestone in understanding placental function came in 1774 with William Hunter's seminal work, The Anatomy of the Human Gravid Uterus Exhibited in Figures, which provided detailed anatomical illustrations based on dissections of pregnant uteri. Hunter demonstrated the separate maternal and fetal vascular systems within the placenta, refuting earlier notions of direct blood mixing and establishing it as a barrier-mediated exchange organ essential for fetal nourishment and waste removal.[154] This anatomical clarity advanced obstetrics by clarifying circulatory dynamics, paving the way for improved surgical and diagnostic approaches in the centuries that followed.[155]The 20th century marked a transformative shift in placental management during the third stage of labor, transitioning from passive expectant approaches to active intervention protocols. Introduced widely in the mid-1900s and endorsed by organizations like the World Health Organization, active management—involving prophylactic oxytocin administration, controlled cord traction, and uterine massage—reduced the incidence of postpartum hemorrhage due to uterine atony by approximately 60%, a primary cause of maternal mortality.[156] This evidence-based strategy contributed to a broader 90% decline in overall maternal mortality from hemorrhage in high-resource settings over the century, through combined advances in hygiene, transfusion, and obstetric care.[157]In the 2020s, telemedicine has expanded remote monitoring of placental function in high-risk pregnancies, particularly in global health contexts with limited access to in-person care. Programs utilizing wearable devices and home-based ultrasound enable real-time assessment of parameters like fetal heart rate and placental blood flow, reducing unnecessary hospitalizations and improving outcomes in underserved regions.[158] Concurrently, ethical debates have intensified around placental tissue use for research and donation, with post-2000 regulations—such as those from the International Society for Stem Cell Research, including 2021 revisions emphasizing equity and access—mandating distinct, informed consent processes to ensure voluntariness and transparency, addressing concerns over commercialization and privacy in stem cell and regenerative applications.[159][160]